EP2528894A1 - Verfahren zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen daraus - Google Patents

Verfahren zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen daraus

Info

Publication number
EP2528894A1
EP2528894A1 EP11703939A EP11703939A EP2528894A1 EP 2528894 A1 EP2528894 A1 EP 2528894A1 EP 11703939 A EP11703939 A EP 11703939A EP 11703939 A EP11703939 A EP 11703939A EP 2528894 A1 EP2528894 A1 EP 2528894A1
Authority
EP
European Patent Office
Prior art keywords
formula
compound
trifluoromethyl
cyclopentyl
benzyloxy
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP11703939A
Other languages
English (en)
French (fr)
Inventor
Antonio Garrido Montalban
Daniel John Buzard
John Aldo Demattei
Tawfik Gharbaoui
Stephen Robert Johannsen
Ashwin M. Krishnan
Young Mi Kuhlman
You-An Ma
Michael John Martinelli
Suzanne Michiko Sato
Dipanjan Sengupta
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Arena Pharmaceuticals Inc
Original Assignee
Arena Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Arena Pharmaceuticals Inc filed Critical Arena Pharmaceuticals Inc
Priority to EP22193921.8A priority Critical patent/EP4148045A1/de
Priority to EP18156371.9A priority patent/EP3378854B1/de
Publication of EP2528894A1 publication Critical patent/EP2528894A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C17/00Preparation of halogenated hydrocarbons
    • C07C17/26Preparation of halogenated hydrocarbons by reactions involving an increase in the number of carbon atoms in the skeleton
    • C07C17/272Preparation of halogenated hydrocarbons by reactions involving an increase in the number of carbon atoms in the skeleton by addition reactions
    • C07C17/278Preparation of halogenated hydrocarbons by reactions involving an increase in the number of carbon atoms in the skeleton by addition reactions of only halogenated hydrocarbons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • C07D209/82Carbazoles; Hydrogenated carbazoles
    • C07D209/88Carbazoles; Hydrogenated carbazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the ring system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • C07D209/94[b, c]- or [b, d]-condensed containing carbocyclic rings other than six-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/14Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D295/155Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms separated by carbocyclic rings or by carbon chains interrupted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P17/00Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms
    • C12P17/10Nitrogen as only ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • the present invention relates to processes and intermediates useful in the preparation of of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l,2,3,4-tetrahydrocyclopenta[b]indol- 3-yl)acetic acid of Formula (la) or salts thereof, an SlPl receptor modulator that is useful in the treatment of SlPl receptor-associated disorders, for example, diseases and disorders mediated by lymphocytes, transplant rejection, autoimmune diseases and disorders, inflammatory diseases and disorders (e.g. , acute and chronic inflammatory conditions), cancer, and conditions characterized by an underlying defect in vascular integrity or that are associated with angiogenesis such as may be pathologic (e.g. , as may occur in inflammation, tumor development and atherosclerosis).
  • SlPl receptor modulator that is useful in the treatment of SlPl receptor-associated disorders, for example, diseases and disorders mediated by lymphocytes, transplant rejection, autoimmune diseases and disorders, inflammatory diseases and disorders
  • SlPl receptor agonists have been shown to possess at least immunosuppressive, antiinflammatory, and/or hemostatic activities, e.g. by virtue of modulating leukocyte trafficking, sequestering lymphocytes in secondary lymphoid tissues, and/or enhancing vascular integrity. Accordingly, SlPl receptor agonists can be useful as immunosuppressive agents for at least autoimmune diseases and disorders, inflammatory diseases and disorders (e.g. , acute and chronic inflammatory conditions), transplant rejection, cancer, and/or conditions that have an underlying defect in vascular integrity or that are associated with angiogenesis such as may be pathologic (e.g., as may occur in inflammation, tumor development, and atherosclerosis) with fewer side effects such as the impairment of immune responses to systemic infection.
  • autoimmune diseases and disorders e.g. , acute and chronic inflammatory conditions
  • transplant rejection e.g., transplant rejection, cancer, and/or conditions that have an underlying defect in vascular integrity or that are associated with angiogenesis
  • angiogenesis e.
  • the sphingosine-1 -phosphate (SIP) receptors 1-5 constitute a family of G protein- coupled receptors containing a seven-transmembrane domain. These receptors, referred to as SlPl to S1P5 (formerly termed endothelial differentiation gene (EDG) receptor-1, -5, -3, -6, and -8, respectively; Chun et al., Pharmacological Reviews, 54:265-269, 2002), are activated via binding by sphingosine-1 -phosphate, which is produced by the sphingosine kmase-catalyzed phosphorylation of sphingosine.
  • SlPl to S1P5 (formerly termed endothelial differentiation gene (EDG) receptor-1, -5, -3, -6, and -8, respectively; Chun et al., Pharmacological Reviews, 54:265-269, 2002)
  • SlPl, S1P4, and S1P5 receptors activate Gi but not Gq, whereas S1P2 and S1P3 receptors activate both Gi and Gq.
  • (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l , 2,3,4- tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) is useful in the treatment of SlPl receptor-associated disorders, such as, psoriasis and multiple sclerosis, and is disclosed in PCT patent application, Serial No. PCT/US2009/004265 hereby incorporated by reference in its entirety.
  • One aspect of the present invention relates to processes and intermediates that are useful in preparing the L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)ben2yloxy)- l,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid, the salt which was found to be surprisingly and unexpectedly different from what was previously reported in PCT patent application, Serial No. PCT/US2009/004265.
  • the present invention provides, inter alia, processes for preparing an L-arginine salt of (R)-2-(7-(4-cyclopen1yl-3-(triiluoromethyl)benzyloxy)-l ,2,3,4-telTahydrocyclopenta[b]indol-3- yl)acetic acid of Formul
  • LG 1 is selected from the group consisting of CI, Br, I, TfO, and TsO, in the presence of:
  • R 1 and R 2 are each independently Ci-C 6 alkyl, or R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 5-member or 6-member heterocyclic ring, and R 3 is C,-C 6 alkyl;
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, Ci-C 4 alkyl, C1-C4 alkoxy, halogen, C1-C4 haloalkyl, C1-C4 haloalkoxy, and nitro;
  • R 3 is Ci-C 6 alkyl
  • the present invention further provides processes for preparing (R)-2-(7-(4-cyclopentyl- 3-(trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la):
  • R 3 is Ci-C 6 alkyl
  • the present invention further provides processes for preparing a compound of Formula
  • R 3 is Ci-C 6 alkyl
  • the alkylating-step solvent is other than dimethylformamide (DMF), or dimethylacetamide (DMA).
  • the present invention further provides processes for preparing a compound of Formula my wherein R 3 is Ci-C 6 alkyl;
  • R 3 is C C 6 alkyl
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, Ci-C 4 alkyl, Ci-C 4 alkoxy, halogen, Ci-C 4 haloalkyl, C C 4 haloalkoxy, and nitro;
  • the present invention further provides processes for preparing a compound of Formula
  • R 3 is Ci-C 6 alkyl
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, Ci-C 4 alkyl, C C 4 alkoxy, halogen, Ci-C 4 haloalkyl, C C 4 haloalkoxy, and nitro;
  • R 1 and R 2 are each independently Ci-C 6 alkyl, or R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 5 -member or 6-member heterocyclic ring;
  • the present invention further provides processes for preparing 4-(chloromethyl)-l- cyclopentyl-2-(trifluoromethyl)benz comprising the step of reacting l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (lib)): with 1,3,5-trioxane in the presence of an acid and a chlorinating agent, to form 4- (chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene of Formula (He).
  • the present invention further provides processes for preparing l-cyclopentyl-2-
  • LG 1 is selected from the group consisting of CI, Br, I, TfO, and TsO, in the presence of:
  • compositions comprising (R)-2-
  • the present invention further provides processes of preparing a pharmaceutical composition
  • a pharmaceutical composition comprising admixing (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)- l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la), or a salt thereof:
  • the present invention further provides pharmaceutical compositions comprising an L- arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l,2,3,4- tetrahydrocyclopenta[b
  • the present invention further provides processes of preparing a pharmaceutical composition
  • a pharmaceutical composition comprising admixing an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-! ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula
  • the present invention further provides compounds represented by any of the formulae described herein.
  • the present invention further provides compounds represented by any of the formulae described herein for use in a process for preparing a pharmaceutical composition for treating an S 1P1 receptor-associated disorder in an individual.
  • the present invention further provides compounds represented by any of the formulae described herein prepared according to any of the processes described herein.
  • the present invention further provides compounds represented by any of the formulae described herein prepared according to any of the processes described herein, for use in a process for preparing a pharmaceutical composition for treating an S1P1 receptor-associated disorder in an individual.
  • Figure 1 shows the effect of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)- l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid (Compound of Formula (la)) in the Peripheral Lymphocyte Lowering (PLL) Assay after a 1 mg/kg oral dose in BALB/c mice.
  • PLL Peripheral Lymphocyte Lowering
  • Figure 2 shows the effect of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)- l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid (Compound of Formula (la)) in the Peripheral Lymphocyte Lowering (PLL) Assay after a 1 mg/kg oral dose in male Sprague- Dawley rats.
  • PLL Peripheral Lymphocyte Lowering
  • Figure 3 shows the reduction of mean ankle diameter after 0.3 mg/kg, 1 mg/kg, and 3 mg/kg dosing of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l , 2,3,4- tetrahydrocyclopenta[b]indol-3-yl)acetic acid (Compound of Formula (la)) in the female Lewis rat collagen-induced arthritis assay.
  • Figure 4 shows the effect of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)- l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid (Compound of Formula (la)) in the experimental autoimmune encephalomyelitis (EAE) assay after daily oral dosing of 0.3 mg/kg, 1 mg/kg, and 3 mg/kg from day 3 to day 21.
  • EAE experimental autoimmune encephalomyelitis
  • Figure 5 shows a powder X-ray diffraction (PXRD) pattern for the L-arginine salt of
  • Figure 6 shows a differential scanning calorimetry (DSC) thermogram for the L- arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l , 2,3,4- tetrahydrocyclopenta[b]indol-3-yl)acetic acid (Compound of Formula (la)).
  • DSC differential scanning calorimetry
  • Figure 7 shows a thermogravimetric analysis (TGA) thermogram for the L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol- 3-yl)acetic acid (Compound of Formula (la)).
  • TGA thermogravimetric analysis
  • Figure 8 shows a moisture sorption analysis for the L-arginine salt of (R)-2-(7-(4- cyclopentyl-3-(1rifluoromethyl)benzyloxy)-l ,2,3,4-tetTahydrocyclopenta[b]mdol-3-yl)acetic acid (Compound of Formula (la)).
  • C,-C 4 alkoxy is intended to mean a C 1 -C 4 alkyl radical, as defined herein, attached directly to an oxygen atom. Some embodiments are 1 to 3 carbons and some embodiments are 1 or 2 carbons. Examples include methoxy, ethoxy, w-propoxy, isopropoxy, n- butoxy, teri-butoxy, isobutoxy, sec-butoxy and the like.
  • Ci-C 6 alkyl is intended to mean a straight or branched carbon radical containing 1 to 6 carbons. Some embodiments are 1 to 5 carbons, some embodiments are 1 to 4 carbons, some embodiments are 1 to 3 carbons and some embodiments are 1 or 2 carbons.
  • Examples of an alkyl include, but are not limited to, methyl, ethyl, w-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, terZ-butyl, pentyl, isopentyl, tert-pentyl, weo-pentyl, 1 -methylbutyl [i.e.,
  • Ci-C 4 haloalkoxy is intended to mean a Ci-C 4 haloalkyl, as defined herein, which is directly attached to an oxygen atom. Examples include, but are not limited to, difluoromethoxy, trifiuoromethoxy, 2,2,2-trifluoroethoxy, pentafluoroethoxy and the like.
  • (- C haloalkyl) is intended to mean an C C 4 alkyl group, defined herein, wherein the alkyl is substituted with between one halogen up to fully substituted wherein a fully substituted Ci-C 6 haloalkyl can be represented by the formula C TrustL 2n+ i wherein L is a halogen and "n" is 1, 2, 3, or 4.
  • the halogens may be the same or different and selected from the group consisting of fluoro, chloro, bromo or iodo, preferably fluoro.
  • Some embodiments are 1 to 4 carbons, some embodiments are 1 to 3 carbons and some embodiments are 1 or 2 carbons.
  • haloalkyl groups include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, chlorodifluoromethyl, 2,2,2-trifluoroethyl, pentafluoroethyl, and the like.
  • halogen or halo is intended to mean a fluoro, chloro, bromo, or iodo group.
  • nitro is intended to mean a radical of the formula: -N0 2 .
  • C 1 -C4 alkylalcohol is intended to mean a straight or branched carbon alkane containing 1 to 4 carbons wherein one hydrogen has been replaced with an OH group.
  • Examples of a C 1 -C4 alkylalcohol include, but are not limited to, methanol, ethanol, isopropanol, n- butanol, tert-butanol, and the like.
  • agonists is intended to mean moieties that interact and activate a receptor, such as the SlPl receptor, and initiate a physiological or pharmacological response characteristic of that receptor, for example, moieties that activate the intracellular response upon binding to the receptor, or enhance GTP binding to membranes.
  • hydrate means a compound, including but not limited to a pharmaceutically acceptable salt of a compound, that further includes a stoichiometric or non- stoichiometric amount of water bound by non-covalent intermolecular forces.
  • mice rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates and most preferably humans.
  • composition is intended to mean a composition comprising at least one active ingredient; including but not limited to Compound of Formula (la) and pharmaceutically acceptable salts, solvates and hydrates thereof, whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, without limitation, a human).
  • a mammal for example, without limitation, a human
  • Those of ordinary skill in the art will understand and appreciate the techniques appropriate for determining whether an active ingredient has a desired efficacious outcome based upon the needs of the artisan.
  • solvate means a compound, including but not limited to a pharmaceutically acceptable salt of a compound, that further includes a stoichiometric or non- stoichiometric amount of a solvent bound by non-covalent intermolecular forces.
  • Preferred solvents are volatile, non-toxic, and/or acceptable for administration to humans in trace amounts.
  • treatment includes one or more of the following:
  • prevention of a disease for example, prevention of a disease, condition or disorder in an individual that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease;
  • inhibition of a disease for example, inhibition of a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or
  • amelioration of a disease for example, amelioration of a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology).
  • Compound of Formula (la) and pharmaceutically acceptable salts, solvates and hydrates thereof can be used in a protective or preventive manner; or Compound of Formula (la) and pharmaceutically acceptable salts, solvates and hydrates thereof can be used to alleviate, inhibit or ameliorate a disease, condition or disorder.
  • the present invention is directed, inter alia, to processes and intermediates useful in the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l ,2,3,4- tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) and/or salts related thereto.
  • One aspect of the present invention pertains to processes, such as those exemplified by Schemes I, II, ⁇ , IV, V, VI, and VII, (supra), that involve Compounds (la), (Ha), (lib), (lie), (lid), (lie), (Ilf), (Ilg), (Ilh), ( ⁇ ), (Ilj), and (Ilk).
  • One aspect of the present invention pertains to intermediates, Compounds (la), (Ila), (lib), (lie), (lid), (He), (Hi), (Ilg), (Ilh), (Ili), (Ilj), and (Ilk), as exemplified in Schemes I, II, III, IV, V, VI, and VII (supra), useful in the preparation of Compound of Formula (la) and/or a salt related thereto, for example, an L-arginine salt of Compound of formula (la).
  • LG 1 is selected from the group consisting of CI, Br, I, TfO, and TsO;
  • R 1 and R 2 are each independently C C 6 alkyl, or R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 5 -member or 6-member heterocyclic ring;
  • R 3 is C,-C 6 alkyl
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, Q-C, alkyl, C1-C 4 alkoxy, halogen, C1-C 4 haloalkyl, C1-C 4 haloalkoxy, and nitro.
  • LG 1 is TfO or TsO.
  • LG 1 is TfO.
  • LG 1 is selected from the group consisting of CI, Br, and I.
  • LG 1 is Br or I.
  • LG 1 is Br.
  • R 1 and R 2 are each independently C ⁇ -C 6 alkyl.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 5-member or 6-member heterocyclic ring.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 6-member heterocyclic ring.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 5-member heterocyclic ring.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form a morpholinyl ring. In some embodiments, R 1 and R 2 together with the nitrogen atom to which they are both bonded form a pyrrolidinyl ring.
  • R 3 is methyl or ethyl.
  • R 3 is ethyl
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, CH 3 , OCH 3 , OCH 2 CH 3 , F, CI, Br, CF 3 , OCF 3 , and nitro.
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, CH 3 , OCH 3 , OCH 2 CH 3 , F, CI, CF 3 , OCF 3 .
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H and OCH 3 .
  • R 4 , R 5 , and R 6 are each H.
  • One aspect of the present invention pertains to a compound of Formula (Ili):
  • R 3 is Ci-C 6 alkyl. In some embodiments, R 3 is ethyl.
  • One aspect of the present invention pertains to processes for preparing l-cyclopentyl-2- (trifluoromethyl)benzene of Formula (lib): comprising the step of cross-coupling bromopentane with a compound of Formula (Ila):
  • LG 1 is selected from the group consisting of CI, Br, I, TfO, and TsO, in the presence of:
  • LG 1 is TfO or TsO.
  • LG 1 is TfO.
  • LG 1 is selected from the group consisting of CI, Br, and I.
  • LG 1 is Br or I.
  • LG 1 is Br.
  • the elemental magnesium is in the form of magnesium turnings, magnesium ribbons, magnesium powder, or magnesium rods.
  • the elemental magnesium is in the form of magnesium turnings.
  • the Fe catalyst is an Fe(ni) catalyst (i.e., Fe +3 ).
  • the Fe catalyst comprises FeF 3 , FeF 3 -3H 2 0, FeCl 3 , FeCl3-6H 2 0,
  • Fe(acac) 3 i.e., Fe(CH 3 C0CHC0CH 3 ) 3
  • Fe(salen)Cl complex has the following formula:
  • the Fe catalyst comprises FeCl 3 .
  • the cross-coupling-step solvent comprises any suitable solvent. In some embodiments, the cross-coupling-step solvent comprises an ethereal solvent. In some embodiments, the cross-coupling-step solvent comprises tetrahydrofuran
  • THF 2-methyl-tetrahydrofuran
  • diethyl ether diethyl ether
  • dibutyl ether diethyl ether
  • te -butylmethyl ether diethyl ether
  • tetrahydropyran 2-methyl-tetrahydrofuran
  • the cross-coupling-step solvent comprises tetrahydrofuran (THF).
  • the cross-coupling agent comprises dimethylacetamide (DMA), dimethylformamide (DMF), N-methylpyrrolidinone ( ⁇ ), hexamethylphosphoric acid triamide (HMPA), l,3-dimethyl-3,4,5,6-tetrahydro-2(lH)-pyrimidinone (DMPU), or NjrjTJf- tetramethylethylenediamine (TMEDA) .
  • DMA dimethylacetamide
  • DMF dimethylformamide
  • N-methylpyrrolidinone
  • HMPA hexamethylphosphoric acid triamide
  • DMPU l,3-dimethyl-3,4,5,6-tetrahydro-2(lH)-pyrimidinone
  • TEDA NjrjTJf- tetramethylethylenediamine
  • the cross-coupling agent comprises N fl'JV'- tetramethylethylenediamine (TMEDA) .
  • the cross-coupling-step solvent comprises tetrahydrofuran (THF) and the cross-coupling agent comprises N,N,N',N'-tetramethylethylenediamine (TMEDA).
  • THF tetrahydrofuran
  • TEDA N,N,N',N'-tetramethylethylenediamine
  • the cross-coupling-step solvent is substantially free of water. In some embodiments, the cross-coupling agent is substantially free of water.
  • the step of cross-coupling bromopentane with a compound of Formula (Ha) is conducted under a substantially inert atmosphere.
  • the step of cross-coupling bromopentane with a compound of Formula (Ha) is conducted under a substantially inert atmosphere comprising argon or nitrogen.
  • Formula (Ha) is conducted under a substantially inert atmosphere comprising nitrogen.
  • the molar ratio between bromopentane and the compound of Formula (Ha) is about 1.0:1.0 to about 10.0:1.0.
  • the molar ratio between bromopentane and the compound of Formula (Ha) is about 1.0:1.0 to about 5.0:1.0.
  • the molar ratio between bromopentane and the compound of Formula (Ila) is about 1.0: 1.0 to about 2.0: 1.0.
  • the molar ratio between bromopentane and the compound of Formula (Ha) is about 1.2:1.0.
  • Fe catalyst is about 1.0:0.01 to about 1.0:1.0.
  • the molar ratio between the compound of Formula (Ila) and the Fe catalyst is about 1.0:0.05 to about 1.0:0.5.
  • the molar ratio between the compound of Formula (Ha) and the Fe catalyst is about 1.0:0.10 to about 1.0:0.3.
  • the molar ratio between the compound of Formula (Ila) and the Fe catalyst is about 1.0:0.15. In some embodiments, the molar ratio between the compound of Formula (Ila) and the elemental magnesium is about 1.0:1.0 to about 1.0:5.0.
  • the molar ratio between the compound of Formula (Ila) and the elemental magnesium is about 1.0:1.0 to about 1.0:3.0.
  • the molar ratio between the compound of Formula (Ila) and the elemental magnesium is about 1.0:1.0 to about 1.0:2.5.
  • the molar ratio between the compound of Formula (Ila) and the elemental magnesium is about 1.0:1.5.
  • the molar ratio between bromopentane, the compound of Formula (Ha), the elemental magnesium, and the Fe catalyst is about 1.2:1.0:1.5:0.15.
  • Formula (Ha) is conducted at a temperature of about 0 °C to about 75 °C.
  • Formula (Ha) is conducted at a temperature of about 10 °C to about 55 °C.
  • Formula (Ha) is conducted at a temperature of about 10 °C to about 45 °C.
  • Formula (Ha) is conducted by adding the cross-coupling agent to a mixture comprising the elemental magnesium, the Fe catalyst, and the cross-coupling-step solvent to form a first cross- coupling mixture.
  • adding the cross-coupling agent to a mixture comprising the elemental magnesium, the Fe catalyst, and the cross-coupling-step solvent is conducted at a rate so the internal temperature during the addition of the cross-coupling agent to the mixture comprising the elemental magnesium, the Fe catalyst, and the cross-coupling-step solvent is maintained at about 0 °C to about 45 °C.
  • adding the cross-coupling agent to a mixture comprising the elemental magnesium, the Fe catalyst, and the cross-coupling-step solvent is conducted at a rate so the internal temperature during the addition of the cross-coupling agent to the mixture comprising the elemental magnesium, the Fe catalyst, and the cross-coupling-step solvent is maintained at about 10 °C to about 30 °C.
  • adding the cross-coupling agent to a mixture comprising the elemental magnesium, the Fe catalyst, and the cross-coupling-step solvent is conducted at a rate so the internal temperature during the addition of the cross-coupling agent to the mixture comprising the elemental magnesium, the Fe catalyst, and the cross-coupling-step solvent is maintained at about 15 °C to about 25 °C.
  • the first cross-coupling mixture is maintained at a temperature of about 10 °C to about 55 °C. In some embodiments, the first cross-coupling mixture is maintained at a temperature of about 20 °C to about 50 °C.
  • the step of cross-coupling bromopentane with a compound of Formula (Ha) further comprises the step of adding a mixture comprising the bromopentane and the compound of Formula (Ila) to the first cross-coupling mixture to form a second cross- coupling mixture.
  • adding the mixture comprising the bromopentane and the compound of Formula (Ila) to the first cross-coupling mixture is conducted at a rate so the internal temperature during the addition of the mixture comprising the bromopentane and the compound of Formula (Ila) to the first cross-coupling mixture is maintained at about 20 °C to about 35 °C.
  • the mixture comprising the bromopentane and the compound of Formula (Ha) to the first cross-coupling mixture is conducted at a rate so the internal temperature during the addition of the mixture comprising the bromopentane and the compound of Formula (Ila) to the first cross-coupling mixture is maintained at about 25 °C to about 30 °C.
  • the second cross-coupling mixture is maintained at a temperature of about 20 °C to about 35 °C.
  • the second cross-coupling mixture is maintained at a temperature of about 20 °C to about 30 °C.
  • the second cross-coupling mixture is maintained at a temperature of about 23 °C to about 27 °C.
  • the step of cross-coupling bromopentane with a compound of Formula (Ha) further comprises the step of quenching the second cross-coupling mixture with aqueous HC1.
  • the elemental magnesium is in the form of magnesium turnings;
  • the Fe catalyst is FeCl 3 ;
  • the cross-coupling-step solvent comprises tetrahydrofuran (THF); and the cross-coupling agent comprises N.NN' ⁇ V'-tetramethylethylenediamine (TMEDA).
  • the molar ratio between bromopentane and the compound of Formula (Ha) is about 1.0: 1.0 to about 2.0: 1.0.
  • the step of cross-coupling bromopentane with a compound of Formula (Ha) is performed wherein the molar ratio between the compound of Formula (Ila) and the Fe catalyst is about 1.0:0.10 to about 1.0:0.3.
  • the step of cross-coupling bromopentane with a compound of Formula (Ha) is performed wherein the molar ratio between the compound of Formula (Ila) and the elemental magnesium is about 1.0: 1.0 to about 1.0:2.5.
  • the step of cross-coupling bromopentane with a compound of Formula (Ha) is performed wherein:
  • the elemental magnesium is in the form of magnesium turnings;
  • the Fe catalyst is FeCl 3 ;
  • the cross-coupling-step solvent comprises tetrahydrofuran (THF); and the cross-coupling agent comprises N,N,N',N'-tetramefoylethylenediamine (TMEDA); and
  • the molar ratio between bromopentane and the compound of Formula (Ila) is about 1.0: 1.0 to about 2.0: 1.0;
  • the molar ratio between the compound of Formula (Ila) and the Fe catalyst is about 1.0:0.10 to about 1.0:0.3;
  • the molar ratio between the compound of Formula (Ila) and the elemental magnesium is about 1.0: 1.0 to about 1.0:2.5.
  • One aspect of the present invention pertains to processes for preparing 4- (chloromethyl)-l-cyclopentyl-2-(triflu a (He):
  • the acid comprises sulfuric acid, acetic acid, trifluoroacetic acid, or methanesulfonic acid.
  • the acid comprises trifluoroacetic acid.
  • the acid comprises sulfuric acid.
  • the chlorinating agent comprises thionyl chloride, oxalyl chloride, phosphorous trichloride, phosphorous pentachloride, phosphorous oxychloride, or chlorosulfonic acid.
  • the chlorinating agent comprises chlorosulfonic acid.
  • the chlorinating agent comprises thionyl chloride.
  • the step of reacting l-cyclopentyl-2-(trifluoromethyl)benzene (Formula lib) with 1,3,5-trioxane in the presence of an acid and a chlorinating agent is conducted in the presence of a suitable solvent.
  • the step of reacting l-cyclopentyl-2-(trifluoromethyl)benzene
  • the step of reacting l-cyclopentyl-2-(trifluoromethyl)benzene (Formula lib) with 1,3,5-trioxane in the presence of an acid and a chlorinating agent is conducted under a substantially inert atmosphere comprising argon or nitrogen.
  • the step of reacting l-cyclopentyl-2-(trifluoromethyl)benzene (Formula lib) with 1,3,5-trioxane in the presence of an acid and a chlorinating agent is conducted under an atmosphere comprising substantially nitrogen.
  • the molar ratio between the l-cyclopentyl-2- (trifluoromethyl)benzene (Formula (Hb)), the 1 ,3,5-trioxane, and the chlorinating agent is about 1.0:0.3:1.0 to about 1.0:3.0:3.0.
  • the molar ratio between the 1 -cyclopentyl-2- (trifluoromethyl)benzene (Formula (lib)), the 1,3,5-trioxane, and the chlorinating agent is about 1.0:1.0:1.5 to about 1.0:2.0:2.5.
  • the molar ratio between the l-cyclopentyl-2- (trifluoromethyl)benzene (Formula (lib)), the 1,3,5-trioxane, the chlorinating agent, and the acid is about 1.0: 1.5:2.0:8.0.
  • the step of reacting l-cyclopentyl-2-(trifluoromethyl)benzene (Formula lib) with 1,3,5-trioxane in the presence of an acid and a chlorinating agent is conducted at a temperature of about -10 °C to about 25 °C.
  • the step of reacting l-cyclopentyl-2-(trifluoromethyl)benzene (Formula lib) with 1,3,5-trioxane in the presence of an acid and a chlorinating agent is conducted at a temperature of about -5 °C to about 15 °C.
  • the step of reacting l-cyclopentyl-2-(trifluoromethyl)benzene
  • the adding the l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (lib)) to the mixture comprising the acid, the chlorinating agent, and the 1,3,5- trioxane is conducted at a rate so the internal temperature during the addition of the 1- cyclopentyl-2-(trifluoromethyl)benzene (Formula (lib)) to the mixture comprising the acid, the chlorinating agent, and the 1,3,5-trioxane is maintained at about -25 °C to about 15 °C.
  • the adding the l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (lib)) to the mixture comprising the acid, the chlorinating agent, and the 1,3,5- trioxane is conducted at a rate so the internal temperature during the addition of the 1- cyclopentyl-2-(trifluoromethyl)benzene (Formula (lib)) to the mixture comprising the acid, the chlorinating agent, and the 1,3,5-trioxane is maintained at about -15 °C to about 10 °C.
  • the adding the l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (lib)) to the mixture comprising the acid, the chlorinating agent, and the 1,3,5- trioxane is conducted at a rate so the internal temperature during the addition of the 1- cyclopentyl-2-(trifluoromethyl)benzene (Formula (lib)) to the mixture comprising the acid, the chlorinating agent, and the 1,3,5-trioxane is maintained at about -10 °C to about 0 °C.
  • the acid comprises sulfuric acid
  • the chlorinating agent comprises thionyl chloride.
  • One aspect of the present invention pertains to processes for preparing a compound of Formula (Hi):
  • R 3 is Ci-C 6 alkyl
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, C C 4 alkyl, C C 4 alkoxy, halogen, Q-C 4 haloalkyl, C 1 -C4 haloalkoxy, and nitro;
  • R 1 and R 2 are each independently Ci-C 6 alkyl, or R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 5 -member or 6-member heterocyclic ring;
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid optionally comprises a drying agent.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid comprises a drying agent.
  • the drying agent is selected from the group of magnesium sulfate, sodium sulfate, and molecular sieves.
  • the drying agent is magnesium sulfate.
  • the drying agent is sodium sulfate.
  • the drying agent is molecular sieves.
  • R 1 and R 2 are each independently Q-Ce alkyl.
  • R 1 and R 2 are each independently methyl or ethyl.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 5-member or 6-member heterocyclic ring. In some embodiments, R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 5 -member heterocyclic ring.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form pyrrolidinyl.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 6-member heterocyclic ring.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form piperidinyl or morpholinyl.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form piperidinyl.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form morpholinyl.
  • R 3 is methyl or ethyl.
  • R 3 is ethyl
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, CH 3 , OCH 3 , OCH 2 CH 3 , F, CI, Br, CF 3 , OCF 3 , and nitro.
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, CH 3 , OCH 3 , OCH 2 CH 3 , F, CI, CF 3 , OCF 3 .
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H and OCH 3 .
  • R 4 , R 5 , and R 6 are each H.
  • the compound of Formula ( ⁇ ) is:
  • the compound of Formula (Ilg) ' is a compound of Formula (Ilg) '
  • the compound of Formula (Ilh) is (4-(benzyloxy)phenyl) hydrazine: , or a salt thereof.
  • the compound of Formula (Ilh) is (4-(benzyloxy)phenyl) hydrazine hydrochloride.
  • the indole-forming acid comprises a Bransted acid or a Lewis acid.
  • the indole-forming acid comprises acetic acid, trifluoroacetic acid, />-TsOH, H 3 P0 4 , H 2 S0 4 , methanesulfonic acid, formic acid, HCl, ZnCl 2 , FeCl 3 , HCl, CuCl, Cul, BF 3 OEt 2 , Zn(Tf) 2 , Yb(Tf) 2 , Sc(Tf) 2 , or A1C1 3 .
  • the indole-forming acid comprises a Bransted acid.
  • the indole-forming acid comprises acetic acid, trifluoroacetic acid, /7-TsOH, H 3 P0 4 , H 2 S0 4 , methanesulfonic acid, formic acid, or HCl. It is understood p- TsOH is para toluenesulfonic acid (i.e., 4-toluenesulfonic acid).
  • the indole-forming acid comprises acetic acid.
  • the indole-forming acid comprises trifluoroacetic acid.
  • the indole-forming acid comprises a mixture of acetic acid and trifluoroacetic acid.
  • the indole-forming acid comprises a Lewis acid.
  • the indole-forming acid comprises ZnCl 2 , FeCl 3 , HCl, CuCl, Cul, BF 3 OEt 2 , Zn(Tf) 2 , Yb(Tf) 2 , Sc(Tf) 2 , or A1C1 3 .
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted in the presence of a suitable solvent.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted in the presence of a protic solvent, a halogenated solvent, an ether solvent, or an aprotic solvent.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted in the presence of a protic solvent.
  • the protic solvent comprises a Ci-C 4 alkylalcohol solvent.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted in the presence of a halogenated solvent.
  • the halogenated solvent comprises dichloromethane.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted in the presence of an ether solvent.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted in the presence of an aprotic solvent.
  • the aprotic solvent comprises acetonitrile or toluene.
  • the aprotic solvent comprises acetonitrile.
  • the aprotic solvent comprises toluene.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted in the presence of a Ci-C 4 alkylalcohol solvent.
  • the Ci-C 4 alkylalcohol solvent comprises methanol or ethanol.
  • the Ci-C 4 alkylalcohol solvent comprises ethanol.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted under a substantially inert atmosphere.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted under a substantially inert atmosphere comprising argon or nitrogen.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted under a substantially inert atmosphere comprising nitrogen.
  • the molar ratio between the compound of Formula (Ilg) and the compound of Formula (Ilh) or a salt thereof is about 1.0: 1.0 to about 1.0:2.0.
  • the molar ratio between the compound of Formula (Ilg) and the compound of Formula (Ilh) or a salt thereof is about 1.0: 1.0 to about 1.0: 1.5.
  • the molar ratio between the compound of Formula (Ilg) and the compound of Formula (Ilh) or a salt thereof is about 1.0: 1.0 to about 1.0: 1.3.
  • the molar ratio between the compound of Formula (Ilg) and the compound of Formula (Ilh) or a salt thereof is about 1.0 : 1.0 to about 1.0: 1.1.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted at a temperature of about 25 °C to about 80 °C.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted at a temperature of about 50 °C to about 70 °C.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is conducted at a temperature of about 60 °C to about 65 °C.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid comprises formation of an imine intermediate of Formula ( ⁇ ):
  • R 3 is ethyl
  • R 4 , R 5 , and R 6 are each H.
  • R 3 is ethyl
  • R 4 , R 5 , and R 6 are each H.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid comprises formation of an imine intermediate of the formula:
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is continued until about 8.0% or less of the compound of Formula ( ⁇ ) is present as determined by
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is continued until about 6.0% or less of the compound of Formula ( ⁇ ) is present as determined by
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is continued until about 5.0% or less of the compound of Formula ( ⁇ ) is present as determined by
  • the step of reacting a compound of Formula (Hg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid is continued until about 4.0% or less of the compound of Formula ( ⁇ ) is present as determined by HPLC.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid further comprises the steps of isomerizing and crystallizing the compound of Formula (Hi) at a temperature of about 20 °C to about 25 °C to form a suspension comprising said compound of Formula (Hi).
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid further comprises the step of cooling said suspension comprising said compound of Formula (Hi) to a temperature of about 0 °C to about 5 °C.
  • the step of reacting a compound of Formula (Ilg) with a compound of Formula (Ilh) or a salt thereof, in the presence of an indole-forming acid further comprises the step of isolating the compound of Formula (Hi).
  • isolating the compound of Formula (Hi) is conducted by filtration.
  • One aspect of the present invention pertains to processes for preparing a compound of Formula Formula (Hg):
  • R 3 is Ci-C 6 alkyl
  • R 1 and R 2 are each independently C 1 -C6 alkyl, or R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 5-member or 6-member heterocyclic ring; to form a compound of Formula (lie):
  • R 3 is C]-C 6 alkyl
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 5-member or 6-member heterocyclic ring.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 5-member heterocyclic ring.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form pyrrolidinyl.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form a 6-member heterocyclic ring.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form piperidinyl or morpholinyl.
  • R 1 and R 2 together with the nitrogen atom to which they are both bonded form morpholinyl.
  • the compound of Formula (lie) is:
  • the step of reacting cyclopentanone with a secondary amine of Formula (lid) is conducted in the presence of an azeotropic solvent.
  • the azeotropic solvent comprises benzene, toluene, cyclohexane or anisole.
  • the azeotropic solvent comprises cyclohexane.
  • the step of reacting cyclopentanone with a secondary amine of Formula (lid) is conducted under a substantially inert atmosphere.
  • the step of reacting cyclopentanone with a secondary amine of Formula (lid) is conducted under an atmosphere comprising argon or nitrogen. In some embodiments, the step of reacting cyclopentanone with a secondary amine of
  • Formula (lid) is conducted under an atmosphere comprising nitrogen.
  • the molar ratio of cyclopentanone and the secondary amine of Formula (Hd) is about 1.0: 1.0 to about 1.0:2.0.
  • Formula (Hd) is about 1.0:1.0 to about 1.0: 1.5.
  • the molar ratio of cyclopentanone and the secondary amine of Formula (Hd) is about 1.0:1.0 to about 1.0: 1.2.
  • the molar ratio of cyclopentanone and the secondary amine of Formula (lid) is about 1.0: 1.0 to about 1.0: 1.05.
  • the molar ratio of cyclopentanone and the secondary amine of Formula (Hd) is about 1.0:1.0 to about 1.0: 1.005.
  • the step of reacting cyclopentanone with a secondary amine of Formula (Hd) is conducted at a temperature of about 60 °C to about 155 °C.
  • Formula (lid) is conducted at a temperature of about 65 °C to about 111 °C.
  • the step of reacting cyclopentanone with a secondary amine of Formula (lid) is conducted at a temperature of about 85 °C to about 95 °C.
  • the step of reacting cyclopentanone with a secondary amine of Formula (Hd) further comprises a step of removing water.
  • the step of reacting cyclopentanone with a secondary amine of Formula (Hd) further comprises a step of removing water via a Dean-Stark water trap.
  • the removing water step is conducted until about 10% or less of cyclopentanone is present as determined by gas chromatography.
  • the removing water step is conducted until about 6% or less of cyclopentanone is present as determined by gas chromatography.
  • the removing water step is conducted until about 3% or less of cyclopentanone is present as determined by gas chromatography.
  • the removing water is conducted until about 10% or less of the secondary amine of Formula (Hd) is present as determined by gas chromatography.
  • the removing water is conducted until about 6% or less of the secondary amine of Formula (Hd) is present as determined by gas chromatography.
  • the removing water is conducted until about 3% or less of the secondary amine of Formula (Hd) is present as determined by gas chromatography.
  • Step b) the step of reacting the compound of Formula (lie) with a compound of Formula (Ilf) is conducted in the presence of an azeotropic solvent.
  • the azeotropic solvent comprises benzene, toluene, cyclohexane or anisole.
  • the azeotropic solvent comprises cyclohexane.
  • the step of reacting the compound of Formula (He) with a compound of Formula (Ilf) is conducted under a substantially inert atmosphere.
  • the step of reacting the compound of Formula (lie) with a compound of Formula (Ilf) is conducted under an atmosphere comprising argon or nitrogen.
  • the step of reacting the compound of Formula (He) with a compound of Formula (Ilf) is conducted under an atmosphere comprising nitrogen.
  • the molar ratio between the compound of Formula (lie) and the compound of Formula (Ilf) is about 1.0:1.0 to about 1.0:2.0.
  • the molar ratio between the compound of Formula (lie) and the compound of Formula (Ilf) is about 1.0:1.0 to about 1.0:1.8.
  • the molar ratio between the compound of Formula (lie) and the compound of Formula (Ilf) is about 1.0:1.0 to about 1.0:1.4.
  • the molar ratio between the compound of Formula (lie) and the compound of Formula (Ilf) is about 1.0:1.0 to about 1.0:1.2.
  • the molar ratio between the compound of Formula (lie) and the compound of Formula (Ilf) is about 1.0:1.1.
  • the step of reacting the compound of Formula (lie) with a compound of Formula (III) is conducted at a temperature of about 25 °C to about 105 °C.
  • the step of reacting the compound of Formula (He) with a compound of Formula (Ilf) is conducted at a temperature of about 55 °C to about 100 °C.
  • the step of reacting the compound of Formula (lie) with a compound of Formula (Ilf) is conducted at a temperature of about 60 °C to about 95 °C.
  • the step of reacting the compound of Formula (lie) with a compound of Formula (Ilf) further comprises a step of removing water.
  • the step of reacting the compound of Formula (lie) with a compound of Formula (Ilf) further comprises a step of removing water via a Dean-Stark water trap.
  • removing water step is continued until about 5.0% or less of the compound of Formula (He) is present as determined by gas chromatography.
  • removing water step is continued until about 2.5% or less of the compound of Formula (He) is present as determined by gas chromatography. In some embodiments, removing water step is continued until about 2.0% or less of the compound of Formula (He) is present as determined by gas chromatography.
  • removing water step is continued until about 1.0% or less of the compound of Formula (He) is present as determined by gas chromatography.
  • removing water step is continued until about 0.5% or less of the compound of Formula (He) is present as determined by gas chromatography.
  • One aspect of the present invention pertains to processes for preparing a compound of Formula (Dj) or a salt thereof:
  • R 3 is Cj-C6 alkyl
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, Ci-C 4 alkyl, Q-C4 alkoxy, halogen, C 1 -C4 haloalkyl, C 1 -C4 haloalkoxy, and nitro;
  • the compound of Formula (Ilj) is: , or a salt thereof, r
  • R 3 is methyl or ethyl.
  • R 3 is ethyl
  • the reducing-step agent comprises formic acid and a reducing base.
  • the reducing base comprises an inorganic base.
  • the reducing base comprises a carbonate base. In some embodiments, the reducing base comprises sodium carbonate, potassium carbonate, or cesium carbonate.
  • the reducing base comprises sodium carbonate.
  • the reducing base comprises potassium carbonate.
  • the reducing base comprises an organic amine base.
  • the reducing base comprises ammonia, dimethylamine, diethylamine, trimethylamine, or triethylamine.
  • the reducing base comprises triethylamine.
  • the reducing-step catalyst comprises palladium.
  • the reducing-step catalyst comprises palladium on carbon.
  • the reducing-step catalyst comprises about 2% palladium on carbon to about 10% palladium on carbon.
  • the reducing-step catalyst comprises about 10% palladium on carbon.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted in the presence of a suitable solvent.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted in the presence of a reducing- step solvent.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted in the presence of a reducing- step solvent comprising a C C 4 alkylalcohol.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted in the presence of a reducing- step solvent comprising methanol or ethanol.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted in the presence of a reducing- step solvent comprising ethyl acetate.
  • the ethyl acetate is substantially free of dissolved oxygen.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted under a substantially inert atmosphere.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted under a substantially inert atmosphere comprising argon or nitrogen. In some embodiments, the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted under a substantially inert atmosphere comprising nitrogen.
  • the molar ratio between the compound of Formula (Ili), formic acid, and the reducing base is about 1.0:1.0:1.0 to about 1.0:6.0:6.0.
  • the molar ratio between the compound of Formula (Ili), formic acid, and the reducing base is about 1.0:1.0:1.0 to about 1.0:5.0:5.0.
  • the molar ratio between the compound of Formula (Hi), formic acid, and the reducing base is about 1.0:2.0:2.0 to about 1.0:4.0:4.0.
  • the molar ratio between the compound of Formula (Ili), formic acid, and the reducing base is about 1.0:2.0:2.0 to about 1.0:3.0:3.0.
  • the molar ratio between the compound of Formula (Ili), formic acid, and the reducing base is about 1.0:3.0:3.0.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted at a temperature of about 15 °C to about 55 °C.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted at a temperature of about 20 °C to about 45 °C.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted at a temperature of about 25 °C to about 35 °C.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted by adding the reducing base to a mixture comprising the compound of Formula (Hi), formic acid, the reducing-step catalyst, and the reducing-step solvent.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst is conducted by adding triethylamine to a mixture comprising the compound of Formula (Hi), formic acid, the reducing-step catalyst, and the reducing-step solvent at a temperature of about 25 °C to about 35 °C, wherein the compound of Formula (Hi) is:
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a reducing-step catalyst further comprises the step of crystallizing the compound of Formula (Ilj) in the presence of ethyl acetate and heptanes.
  • crystallizing the compound of Formula (Ilj) is conducted at a temperature of about 0 °C to about 20 °C.
  • crystallizing the compound of Formula (Ilj) is conducted at a temperature of about 5 °C to about 15 °C.
  • crystallizing the compound of Formula (Ilj) is conducted at a temperature of about 10 °C.
  • the step of reducing a compound of Formula (Hi) in the presence of a reducing-step agent, and a rediicing-step catalyst further comprises the step of isolating the compound of Formula (Ilj).
  • isolating the compound of Formula (Ilj) is conducted by filtration.
  • One aspect of the present invention pertains to processes for preparing a compound of Formula (Ilk):
  • R 3 is Ci-C 6 alkyl
  • the alkylating-step solvent is other than dimethylformamide
  • the alkylating-step solvent is other than dimethylacetamide
  • the alkylating-step solvent is other than a solvent of the group consisting of dimethylformamide (DMF) and dimethylacetamide (DMA).
  • the compound of Formula (Ilk) is:
  • R 3 is methyl or ethyl.
  • R 3 is ethyl
  • the alkylating-step base is an inorganic base.
  • the alkylating-step base comprises a carbonate base.
  • the alkylating-step base comprises sodium carbonate, potassium carbonate, or cesium carbonate.
  • the alkylating-step base comprises cesium carbonate.
  • the alkylating-step solvent comprises a suitable solvent.
  • the alkylating-step solvent comprises an aprotic solvent.
  • the alkylating-step solvent comprises acetone, 2-butanone, dimethylformamide (DMF), dimethylacetamide (DMA), tetrahydrofuran (THF) or acetonitrile.
  • the alkylating-step solvent comprises acetonitrile.
  • the alkylating-step solvent is substantially free of water.
  • the step of alkylating a compound of Formula (Dj) or a salt thereof, with 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (He)) in the presence of an alkylating-step base, and an alkylating-step solvent, is conducted wherein: the compound of Formula (Ilj) is: or a salt thereof;
  • the alkylating-step base comprises cesium carbonate
  • the alkylating-step solvent comprises acetonitrile.
  • the step of alkylating a compound of Formula (Ilj) or a salt thereof, with 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (He)) in the presence of an alkylating-step base, and an alkylating-step solvent is conducted under a substantially inert atmosphere.
  • the step of alkylating a compound of Formula (Dj) or a salt thereof, with 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (He)) in the presence of an alkylating-step base, and an alkylating-step solvent is conducted under a substantially inert atmosphere comprising argon or nitrogen.
  • the step of alkylating a compound of Formula (Ilj) or a salt thereof, with 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (He)) in the presence of an alkylating-step base, and an alkylating-step solvent is conducted under a substantially inert atmosphere comprising nitrogen.
  • the molar ratio between 4-(chloromethyl)-l-cyclopentyl-2- (trifluoromethyl)benzene (Formula (He)), the compound of Formula (Ilj) or a salt thereof, and the alkylating-step base is about 1.0:1.0:0.5 to about 2.0:1.0:3.0.
  • the step of alkylating a compound of Formula (Ilj) or a salt thereof, with 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (He)) in the presence of an alkylating-step base, and an alkylating-step solvent is conducted at a temperature of about 15 °C to about 90 °C.
  • the step of alkylating a compound of Formula (Ilj) or a salt thereof, with 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (He)) in the presence of an alkylating-step base, and an alkylating-step solvent is conducted at a temperature of about 21 °C to about 85 °C.
  • the step of alkylating a compound of Formula (Ilj) or a salt thereof, with 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (lie)) in the presence of an alkylating-step base, and an alkylating-step solvent is conducted at a temperature of about 65 °C to about 80 °C.
  • the step of alkylating a compound of Formula (Ilj) or a salt thereof, with 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (He)) in the presence of an alkylating-step base, and an alkylating-step solvent further comprises the step of adding 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (lie)) to a mixture comprising the compound of Formula (Ilj) or a salt thereof, alkylating-step base, and alkylating- step solvent to form an alkylating mixture.
  • adding 4-(chloromethyl)-l-cyclopentyl-2- (trifluoromethyl)benzene (Formula (He)) to the mixture comprising the compound of Formula (Ilj) or a salt thereof, alkylating-step base, and alkylating-step solvent is conducted with heating so the internal temperature during the addition of 4-(chloromethyl)-l-cyclopentyl-2- (trifluoromethyl)benzene (Formula (lie)) to the alkylating mixture comprising the compound of Formula (Ilj) or a salt thereof, alkylating-step base, and alkylating-step solvent is about 20 °C to about 85 °C.
  • the alkylating mixture is maintained at about 60 °C to about 85
  • the alkylating mixture is maintained at about 70 °C to about 85
  • the alkylating mixture is maintained at about 75 °C to about 80 °C.
  • the step of alkylating a compound of Formula (Ilj) or a salt thereof, with 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (lie)) in the presence of an alkylating-step base, and an alkylating-step solvent further comprises the steps of cooling the alkylating mixture to a temperature of about 50 °C to about 60 °C and filtering the alkylating mixture to form a filtered mixture.
  • the step of alkylating a compound of Formula (Ilj) or a salt thereof, with 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (He)) in the presence of an alkylating-step base, and an alkylating-step solvent further comprises the step of precipitating the compound of Formula (Ilk) from the filtered mixture.
  • precipitating the compound of Formula (Ilk) from the filtered mixture comprises reducing the volume of the filtered mixture.
  • precipitating the compound of Formula (Ilk) from the filtered mixture comprises reducing the volume of the filtered mixture by about one half.
  • the step of alkylating a compound of Formula (Qj) or a salt thereof, with 4-(chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene (Formula (lie)) in the presence of an alkylating-step base, and an alkylating-step solvent further comprises isolating the precipitate of the compound of Formula (Ilk) from the filtered mixture.
  • isolating the precipitate of the compound of Formula (Ilk) is conducted by filtration.
  • R 3 is d-C 6 alkyl
  • R 3 is methyl or ethyl.
  • R 3 is ethyl
  • the lipase is selected from the group consisting of lipase B Candida Antarctica, lipase Mucor miehei, and P. fluorescens.
  • the lipase is Candida antarctica lipase B.
  • the lipase is immobilized Candida antarctica lipase B.
  • the hydrolyzing-step solvent comprises a suitable solvent.
  • the hydrolyzing-step solvent comprises dimethylformamide (DMF), dimethylacetamide (DMA), dimethylsulfoxide (DMSO), tetrahydrofuran (THF), or acetonitrile.
  • the hydrolyzing-step solvent comprises acetonitrile.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted wherein:
  • the lipase is immobilized Candida antarctica lipase B;
  • the hydrolyzing-step solvent comprises acetonitrile.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer at a pH of about 6.0 to about 9.0.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer at a pH of about 7.0 to about 8.5.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer at a pH of about 7.3 to about 8.3.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer at a pH of about 7.6 to about 8.0.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer at a pH of about 7.8.
  • the phosphate buffer is a sodium phosphate buffer.
  • the phosphate buffer is a potassium phosphate buffer.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted at a temperature of about 0 °C to about 75 °C.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted at a temperature of about 20 °C to about 65 °C.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted at a temperature of about 30 °C to about 55 °C. In some embodiments, the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted at a temperature of about 35 °C to about 45 °C.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted at a temperature of about 40 °C.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent further comprises the step of isolating the (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3- yl)acetic acid of Formula (la).
  • (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) has an enantiomeric excess of about 95% or greater.
  • (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) has an enantiomeric excess of about 98% or greater.
  • (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) has an enantiomeric excess of about 99% or greater.
  • One aspect of the present invention pertains to processes for preparing an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(1rifluoromethyl)benzyloxy)-l ,2,3,4-telTahyo 'ocyclopenta[3 ⁇ 4]indol- 3-yl)acetic acid of
  • R 3 is Ci-C 6 alkyl
  • R 3 is methyl or ethyl.
  • R 3 is ethyl
  • the lipase is Candida antarctica lipase B.
  • the lipase is immobilized Candida antarctica lipase B.
  • the hydrolyzing-step solvent comprises a suitable solvent.
  • the hydrolyzing-step solvent comprises dimethylformamide (DMF), dimethylacetamide (DMA), dimethylsulfoxide (DMSO), tetrahydrofuran (THF), or acetonitrile.
  • the hydrolyzing-step solvent comprises acetonitrile.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted wherein:
  • the lipase is immobilized Candida antarctica lipase B;
  • the hydrolyzing-step solvent comprises acetonitrile.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer. In some embodiments, the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer at a pH of about 6.0 to about 9.0.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer at a pH of about 7.0 to about 8.5.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer at a pH of about 7.3 to about 8.3.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer at a pH of about 7.6 to about 8.0.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted in the presence of a phosphate buffer at a pH of about 7.8.
  • the phosphate buffer is a sodium phosphate buffer.
  • the phosphate buffer is a potassium phosphate buffer.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted at a temperature of about 0 °C to about 75 °C.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted at a temperature of about 20 °C to about 65 °C.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted at a temperature of about 30 °C to about 55 °C.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent is conducted at a temperature of about 35 °C to about 45 °C.
  • the step of hydrolyzing the compound of Formula (Ilk) in the presence of a lipase and a hydrolyzing-step solvent further comprises the step of isolating (R)-2- (7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3- yl)acetic acid of Formula (la).
  • (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) has an enantiomeric excess of about 95% or greater.
  • (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) has an enantiomeric excess of about 98% or greater.
  • (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) has an enantiomeric excess of about 99% or greater.
  • HPLC methods that retention times provided herein are approximate and are depend on numerous parameters that are know by those skilled in art, for example, the column, the column temperature, flow rate, solvent(s), the HPLC system,- and the like.
  • a standard for any of the compounds described herein can be readily prepared and the retention time easily determined for a HPLC system and conditions other than those described herein.
  • CCoolluummnn : Chiralpak IB, 5 ⁇ , 4.6 x 250 mm
  • the retention time for (S)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l ,2,3,4- tetrahydrocyclopenta[b]indol-3-yl)acetic acid using the above conditions is about 30.3 minutes.
  • the contacting-step solvent comprises a suitable solvent.
  • the contacting-step solvent comprises a Ci-C 6 alcohol.
  • the contacting-step solvent comprises isopropyl alcohol.
  • the contacting in step b), is conducted under a substantially inert atmosphere.
  • the contacting in step b), is conducted under a substantially inert atmosphere comprising argon or nitrogen.
  • the contacting in step b), is conducted under a substantially inert atmosphere comprising nitrogen.
  • the molar ratio between (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid (Formula (la)) and L-arginine is about 1.0: 1.0 to about 1.0: 1.2.
  • the molar ratio between (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid (Formula (la)) and L-arginine is about 1.0: 1.0.
  • the contacting in step b) further comprises the step of adding an aqueous solution of L-arginine to a first contacting mixture comprising (R)-2-(7-(4-cyclopentyl- 3-(trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid (Formula (la)) and the Ci-C 6 alcohol to form a second contacting mixture.
  • a first contacting mixture comprising (R)-2-(7-(4-cyclopentyl- 3-(trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid (Formula (la)) and the Ci-C 6 alcohol to form a second contacting mixture.
  • the first contacting mixture is at a temperature of about 45 °C to about 75 °C.
  • the first contacting mixture is at a temperature of about 50 °C to about 70 °C.
  • the first contacting mixture is at a temperature of about 55 °C to about 65 °C.
  • the first contacting mixture is at a temperature of about 60 °C. In some embodiments, further comprising the steps of cooling the second contacting mixture and crystallizing the L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-
  • the cooling is conducted at a rate of about 0.04 °C/minute to about 4.0 °C/minute.
  • the cooling is conducted at a rate of about 0.1 °C/minute to about 2.0 °C/minute.
  • the cooling is conducted at a rate of about 0.4 °C/minute to about 1.0 °C/minute.
  • the cooling is conducted at a rate of about 0.4 °C/minute.
  • contacting (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)- 1 ,2,3 ,4-tetrahydrocyclopenta[b]indol-3 -yl)acetic acid (Formula (la)) with L-arginine or a salt thereof, in the presence of a contacting-step solvent and H 2 0, further comprises the step of isolating the L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-
  • the isolating is conducted by filtration.
  • the L-arginine salt of (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) has a purity of about 95% or greater as determined by HPLC.
  • the L-arginine salt of (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)- 1 ,2,3 ,4-tetrahydrocyclopenta[b]indol-3 -yl)acetic acid of Formula (la) has a purity of about 98% or greater as determined by HPLC.
  • the (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) after isolating the L-arginine salt of (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la), the (R)-2-(7-(4-cyclopentyl-3 -(trifluoromethyl)benzyloxy)- 1 ,2,3 ,4- tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) has an enantiomeric excess of about 95% or greater.
  • the (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)- 1 ,2,3 ,4-tetrahydrocyclopenta[b]indol-3 -yl)acetic acid of Formula (la) after isolating the L-arginine salt of (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)- 1 ,2,3 ,4-tetrahydrocyclopenta[b]indol-3 -yl)acetic acid of Formula (la), the (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l , 2,3,4- tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) has an enantiomeric excess of about 98% or greater. In some embodiments, after isolating the L-arginine
  • the retention time of L-arginine present in the L-arginine salt of (R)-2-(7-(4- cyclopentyl-3-(trifluoromemyl)benzyloxy)-l,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid using the above conditions is about 8.6 minutes.
  • the processes described herein can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., ⁇ or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., ⁇ or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), or mass spectrometry
  • chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • preparation of compounds can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in Greene and Wuts, Protective Groups in Organic Synthesis, 3 rd Ed., Wiley & Sons, 1999.
  • Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected.
  • reactions can be carried out in the absence of solvent, such as when at least one of the reagents is a liquid or gas.
  • Suitable solvents can include halogenated solvents such as: carbon tetrachloride, bromodichloromethane, dibromochloromethane, bromoform, chloroform, bromochloromethane, dibromomethane, butyl chloride, dichloromethane, tetrachloroethylene, trichloroethylene, 1,1,1- trichloroethane, 1,1,2-trichloroethane, 1,1-dichloroethane, 2-chloropropane, hexafluorobenzene, 1,2,4-trichlorobenzene, 1 ,2-dichlorobenzene, 1,3-dichlorobenzene, 1,4-dichlorobenzene, chlorobenzene, fluorobenzene, fluorotrichloromethane, chlorotrifluoromethane,
  • halogenated solvents such as: carbon tetrachloride, bromodichloromethane, dibro
  • bromotrifluoromethane carbon tetrafluoride, dichlorofluoromethane, chlorodifiuoromethane, trifluoromethane, 1 ,2-dichlorotetrafluorethane and hexafluoroethane.
  • Suitable solvents can include ether solvents, such as: dimethoxymethane,
  • Suitable solvents can include protic solvents, such as: water, methanol, ethanol, 2- nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, 2-propanol, 2- methoxyethanol, 1-butanol, 2-butanol, isobutyl alcohol, /-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3- pentanol, neo-pentyl alcohol, t-pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.
  • protic solvents such as: water, methanol, ethanol, 2- nitroethanol, 2-fluoroethanol, 2,2,2-trifluoroethanol, ethylene glycol, 1-propanol, 2-propanol, 2- methoxyethanol, 1-butanol, 2-butanol
  • Suitable solvents can include aprotic solvents, such as: benzene, cyclohexane, pentane, hexane, toluene, cycloheptane, methylcyclohexane, heptane, ethylbenzene, o, m-, or ⁇ -xylene, octane, indane, nonane, naphthalene, tetrahydrofuran, acetonitrile, dimethyl sulfoxide, propionitrile, ethyl formate, methyl acetate, hexachloroacetone, acetone, ethyl methyl ketone, ethyl acetate, isopropyl acetate, sulfolane, l,3-dimethyl-3,4,5,6-tetrahydro-2(lH)-pyrimidinone, l,3-dimethyl-2-imidazolidinone,
  • R, R', and R" may be the same or different.
  • R, R', and R" are each independently selected from ⁇ and Ci-C 6 alkyl.
  • R, R', and R" are each independently selected from ⁇ and C 1 -C4 alkyl.
  • R, R', and R" are each independently selected from ⁇ and Ci-C 2 alkyl.
  • Supercritical carbon dioxide can also be used as a solvent.
  • reaction temperatures will depend on, for example, the melting and boiling points of the reagents and solvent, if present; the thermodynamics of the reaction (e.g., vigorously exothermic reactions may need to be carried out at reduced temperatures); and the kinetics of the reaction (e.g., a high activation energy barrier may need elevated temperatures).
  • reactions of the processes described herein can be carried out in air or under an inert atmosphere.
  • reactions containing reagents or products that are substantially reactive with air can be carried out using air-sensitive synthetic techniques that are well known to one skilled in the art.
  • preparation of compounds can involve the addition of acids or bases to effect, for example, catalysis of a desired reaction or formation of salt forms such as acid addition salts.
  • Example acids can be inorganic or organic acids.
  • Inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, and nitric acid.
  • Organic acids include formic acid, acetic acid, trifluoroacetic acid, propionic acid, butanoic acid, methanesulfonic acid,p-toluene sulfonic acid, benzenesulfonic acid, propiolic acid, butyric acid, 2-butynoic acid, vinyl acetic acid, pentanoic acid, hexanoic acid, heptanoic acid, octanoic acid, nonanoic acid and decanoic acid.
  • Example bases include lithium hydroxide, sodium hydroxide, potassium hydroxide, lithium carbonate, sodium carbonate, and potassium carbonate.
  • Some example strong bases include, but are not limited to, hydroxide, alkoxides, metal amides, metal hydrides, metal dialkylamides and arylamines, wherein; alkoxides include lithium, sodium and potassium salts of methyl, ethyl and i-butyl oxides; metal amides include sodium amide, potassium amide and lithium amide; metal hydrides include sodium hydride, potassium hydride and lithium hydride; and metal dialkylamides include sodium and potassium salts of methyl, ethyl, w-propyl, isopropyl, n-butyl, i-butyl, trimethylsilyl and cyclohexyl substituted amides.
  • the compounds described herein can be asymmetric (e.g. , having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Salts of the present invention that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis.
  • the processes described herein can be stereoselective such that any given reaction starting with one or more chiral reagents enriched in one stereoisomer forms a product that is also enriched in one stereoisomer.
  • the reaction can be conducted such that the product of the reaction substantially retains one or more chiral centers present in the starting materials.
  • the reaction can also be conducted such that the product of the reaction contains a chiral center that is substantially inverted relative to a corresponding chiral center present in the starting materials.
  • An example method includes fractional recrystallization (for example, diastereomeric salt resolution) using a "chiral resolving acid" which is an optically active, salt-forming organic acid.
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as ⁇ -camphorsulfonic acid.
  • resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of ⁇ - methylbenzylamine (e.g., S and R forms, or diastereomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, N-methylephedrine, cyclohexylethylamine, 1 ,2-diaminocyclohexane, and the like.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine).
  • Suitable elution solvent composition can be determined by one skilled in the art.
  • the compounds described herein and salts thereof can also include all isotopes of atoms occurring in the intermediates or final compounds or salts thereof. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium.
  • the compounds described herein and salts thereof can also include tautomeric forms, such as keto-enol tautomers. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • the usual isolation and purification operations such as concentration, filtration, extraction, solid-phase extraction, recrystallization, chromatography, and the like may be used, to isolate the desired products.
  • One aspect of the present invention provides, inter alia, intermediates prepared by any of the processes described herein.
  • the present invention further provides pharmaceutical compositions comprising compounds prepared by any of the processes as described herein.
  • the present invention further provides processes of preparing a pharmaceutical composition
  • a pharmaceutical composition comprising admixing Compound of Formula (la) or a salt thereof with a pharmaceutically acceptable carrier, wherein the Compound of Formula (la) or a salt thereof is prepared by any of the processes as described herein.
  • the present invention further provides intermediates, as described herein, for use in processes for preparing pharmaceutical compositions for treating an SIP 1 receptor-associated disorder in an individual.
  • the present invention further provides uses of compounds, as described herein, in processes for preparing pharmaceutical compositions for treating an S1P1 receptor-associated disorder.
  • One aspect of the present invention pertains to compounds represented by any of the formulae described herein.
  • One aspect of the present invention pertains to compounds represented by any of the formulae described herein for use in a process for preparing a pharmaceutical composition for treating an S1P1 receptor-associated disorder in an individual.
  • One aspect of the present invention pertains to compounds represented by any of the formulae described herein prepared according to any of the processes described herein.
  • One aspect of the present invention pertains to compounds represented by any of the formulae described herein prepared according to any of the processes described herein, for use in a process for preparing a pharmaceutical composition for treating an SlPl receptor-associated disorder in an individual.
  • One aspect of the present invention pertains to a compound that is 1 -cyclopentyl-2-
  • One aspect of the present invention pertains to a compound that is l-cyclopentyl-2- (trifluoromethyl)benzene of Formula (lib):
  • One aspect of the present invention pertains to a compound that is l-cyclopentyl-2- (trifluoromethyl)benzene of Formula (lib) prepared according to any of the processes described herein.
  • One aspect of the present invention pertains to a compound that is l-cyclopentyl-2- (trifluoromethyl)benzene of Formula (lib) prepared according to any of the processes described herein, for use in a process for preparing a pharmaceutical composition for treating an SlPl receptor-associated disorder in an individual.
  • One aspect of the present invention pertains to a compound that is 4-(chloromethyl)-l- cyclopentyl-2-(trifluoromethyl)ben
  • One aspect of the present invention pertains to a compound that is 4-(chloromethyl)-l - cyclopentyl-2-(trifluoromethyl)benzene of Formula (lie):
  • One aspect of the present invention pertains to a compound that is 4-(chloromethyl)-l- cyclopentyl-2-(trifluoromethyl)benzene of Formula (lie) prepared according to any of the processes described herein.
  • One aspect of the present invention pertains to a compound that is 4-(chloromethyl)-l- cyclopentyl-2-(trifluoromethyl)benzene of Formula (He) prepared according to any of the processes described herein, for use in a process for preparing a pharmaceutical composition for treating an SlPl receptor-associated disorder in an individual.
  • R 3 is C C 6 alkyl; and R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, C C 4 alkyl, C C 4 alkoxy, halogen, C1-C4 haloalkyl, C1 -C4 haloalkoxy, and nitro.
  • R 3 is ethyl
  • R 4 , R 5 , and R 6 are each H.
  • R 3 is ethyl
  • R 4 , R 5 , and R 6 are each H.
  • R 3 is Ci-C 6 alkyl
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, C C 4 alkyl, C C 4 alkoxy, halogen, C!-C 4 haloalkyl, C C 4 haloalkoxy, and nitro; in the manufacture of a medicament for treating an SlPl receptor-associated disorder.
  • One aspect of the present invention pertains to uses of a compound of the formula:
  • One aspect of the present invention pertains to uses of a compound of Formula (Hi):
  • R 3 is Ci-C 6 alkyl
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, C C 4 alkyl, C]-C 4 alkoxy, halogen, C C 4 haloalkyl, C C 4 haloalkoxy, and nitro; in a process for preparing a pharmaceutical composition for treating an SlPl receptor- associated disorder in an individual.
  • R 3 is C C 6 alkyl
  • R 4 , R 5 , and R 6 are each selected independently from the group consisting of H, Q-C4 alkyl, C 1 -C4 alkoxy, halogen, Ci-C 4 haloalkyl, C 1 -C4 haloalkoxy, and nitro; for use in a process for preparing a pharmaceutical composition for treating an S lPl receptor-associated disorder in an individual.
  • One aspect of the present invention pertains to a compound of Formula (Ili) prepared according to any of the processes described herein.
  • One aspect of the present invention pertains to a compound of Formula (Ili) prepared according to any of the processes described herein, for use in a process for preparing a pharmaceutical composition for treating an S lPl receptor-associated disorder in an individual.
  • One aspect of the present invention pertains to a compound of Formula (Ilj):
  • R 3 is Ci-C 6 alkyl
  • One aspect of the present invention pertains to a compound of Formula (Ilj) prepared according to any of the processes described herein.
  • One aspect of the present invention pertains to a compound of Formula (Ilj) prepared according to any of the processes described herein, for use in a process for preparing a pharmaceutical composition for treating an SlPl receptor-associated disorder in an individual.
  • R 3 is ethyl
  • R 3 is other than ethyl.
  • R 3 is other than methyl.
  • One aspect of the present invention pertains to a compound of Formula (Ilk):
  • R 3 is C]-C 6 alkyl
  • One aspect of the present invention pertains to a compound of Formula (Ilk):
  • R 3 is Ci-C 6 alkyl
  • One aspect of the present invention pertains to a compound of Formula (Ilk) prepared according to any of the processes described herein.
  • One aspect of the present invention pertains to a compound of Formula (Ilk) prepared according to any of the processes described herein, for use in a process for preparing a pharmaceutical composition for treating an S lPl receptor-associated disorder in an individual.
  • R 3 is ethyl
  • R 3 is other than ethyl.
  • R 3 is other than methyl.
  • compositions comprising (R)-2-(7-(4-cyclopen1yl-3-(trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3- yl)acetic acid of Formula (la), or a salt thereof:
  • (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l , 2,3,4- tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) is used for treating an SlPl receptor-associated disorder in an individual, wherein (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) is prepared according to any of the processes described herein.
  • the pharmaceutical composition comprising (R)-2-(7-(4- cyclopentyl-3-(1rifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) is used for treating an S lPl receptor-associated disorder in an individual, wherein (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l ,2,3,4- tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) is prepared according to any of the processes described herein.
  • One aspect of the present invention pertains to processes of preparing a pharmaceutical composition
  • a pharmaceutical composition comprising admixing (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)- l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la), or a salt thereof: and a pharmaceutically acceptable carrier, wherein the (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopentaP5]indol-3-yl)acetic acid of Formula (la) is prepared according to any of the processes described herein.
  • One aspect of the present invention pertains to (R)-2-(7-(4-cyclopentyl-3-
  • One aspect of the present invention pertains to (R)-2-(7-(4-cyclopentyl-3- (trifluoromethyl)benzyloxy)-l ,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) or a salt thereof, prepared according to any of the processes described herein, for use in a process for preparing a pharmaceutical composition for treating an S1P1 receptor-associated disorder in an individual.
  • compositions comprising an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-l ,2,3,4- tetrahydrocyclopenta[b]ind -3-yl)acetic acid of Formula (la):
  • (la) is used for treating an SIP 1 receptor-associated disorder in an individual, wherein the L- arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)ben2yloxy)-l ,2,3,4- tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) is prepared according to any of the processes described herein.
  • the pharmaceutical composition comprising an L-arginine salt of
  • One aspect of the present invention pertains to processes of preparing a pharmaceutical composition comprising admixing an L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-
  • One aspect of the present invention pertains to the L-arginine salt of (R)-2-(7-(4- cyclopentyl-3-(trifluoromemyl)benzyloxy)-l,2,3,4-te1rahydrocyclopenta[3 ⁇ 4]indol-3-yl)acetic acid of Formula (la) or a salt thereof, prepared according to any of the processes described herein.
  • One aspect of the present invention pertains to the L-arginine salt of (R)-2-(7-(4- cyclopentyl-3-(1rifluoromemyl)benzyloxy)-l,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid of Formula (la) or a salt thereof, prepared according to any of the processes described herein, for use in a process for preparing a pharmaceutical composition for treating an S1P1 receptor- associated disorder in an individual.
  • lymphopenia also referred to as peripheral lymphocyte lowering (PLL); Hale et al, Bioorg. Med. Chem. Lett., 14:3351-3355, 2004. This is attended by clinically useful immunosuppression by virtue of sequestering T- and B-cells in secondary lymphoid tissue (lymph nodes and Peyer's patches) and thus apart from sites of inflammation and organ grafts (Rosen et al, Immunol. Rev., 195:160-177, 2003; Schwab et al, Nature
  • lymphocyte sequestration for example in lymph nodes, is thought to be a consequence of concurrent agonist-driven functional antagonism of the S1P1 receptor on T-cells (whereby the ability of SIP to mobilize T-cell egress from lymph nodes is reduced) and persistent agonism of the S1P1 receptor on lymph node endothelium (such that barrier function opposing transmigration of lymphocytes is increased) (Matloubian et al,
  • agonism of the SIP 1 receptor alone is sufficient to achieve lymphocyte sequestration (Sanna et al., J Biol Chem., 279: 13839-13848, 2004) and that this occurs without impairment of immune responses to systemic infection (Brinkmann et al., Transplantation,
  • An exemplary SIP receptor agonist having agonist activity on the S1P1 receptor is FTY720 (fmgolimod), an immunosuppressive agent currently in clinical trials (Martini et al, Expert Opin. Investig. Drugs, 16:505-518, 2007).
  • FTY720 acts as a prodrug which is phosphorylated in vivo; the phosphorylated derivative is an agonist for S1P1, S1P3, S1P4, and S1P5 receptors (but not the S1P2 receptor) (Chiba, Pharmacology & Therapeutics, 108:308- 319, 2005).
  • FTY720 has been shown to rapidly and reversibly induce lymphopenia (also referred to as peripheral lymphocyte lowering (PLL); Hale et al, Bioorg. Med. Chem. Lett., 14:3351-3355, 2004). This is attended by clinically useful immunosuppression by virtue of sequestering T- and B-cells in secondary lymphoid tissue (lymph nodes and Peyer's patches) and thus apart from sites of inflammation and organ grafts (Rosen et al, Immunol. Rev., 195:160-177, 2003; Schwab et al, Nature Immunol, 8:1295-1301, 2007).
  • lymphopenia also referred to as peripheral lymphocyte lowering (PLL); Hale et al, Bioorg. Med. Chem. Lett., 14:3351-3355, 2004. This is attended by clinically useful immunosuppression by virtue of sequestering T- and B-cells in secondary lymphoid tissue (lymph nodes and Pe
  • FTY720 elicited an adverse event (i.e., transient asymptomatic bradycardia) due to its agonism of the S1P3 receptor (Budde et al, J. Am. Soc. Nephrol, 13:1073-1083, 2002; Sanna et al, J. Biol. Chem., 279:13839-13848, 2004; Ogawa et al, BBRC, 361:621-628, 2007).
  • an adverse event i.e., transient asymptomatic bradycardia
  • FTY720 has been reported to have therapeutic efficacy in at least: a rat model for autoimmune myocarditis and a mouse model for acute viral myocarditis (Kiyabayashi et al, J. Cardiovasc. Pharmacol, 35:410-416, 2000; Miyamoto et al, J. Am. Coll. Cardiol, 37:1713- 1718, 2001); mouse models for inflammatory bowel disease including colitis (Mizushima et al, Inflamm. Bowel Dis., 10:182-192, 2004; Deguchi et al, Oncology Reports, 16:699-703, 2006; Fujii et al, Am. J. Physiol. Gastrointest.
  • FTY720 may have therapeutic efficacy for /3-amyloid-related inflammatory diseases including Alzheimer's disease (Kaneider et al, FASEB J., 18:309-311, 2004).
  • KRP-203 an SIP receptor agonist having agonist activity on the S1P1 receptor, has been reported to have therapeutic efficacy in a rat model for autoimmune myocarditis (Ogawa et al, BBRC, 361 :621-628, 2007).
  • SEW2871 agonism of endothelial S1P1 receptors prevents proinflammatory monocyte/endothelial interactions in type I diabetic vascular endothelium (Whetzel et al, Circ. Res., 99:731-739, 2006) and protects the vasculature against TNFa-mediated monocyte/endothelial interactions (Bolick et al, Arterioscler. Thromb. Vase. Biol, 25:976-981, 2005).
  • FTY720 has been reported to have therapeutic efficacy in experimental autoimmune encephalomyelitis (EAE) in rats and mice, a model for human multiple sclerosis (Brinkmann et al, J. Biol. Chem., 277:21453-21457, 2002; Fujino et al, J. Pharmacol. Exp. Ther., 305:70-77, 2003; Webb et al, J. Neuroimmunol, 153: 108-121, 2004; Rausch et al, J. Magn. Reson. Imaging, 20:16-24, 2004; Kataoka et al, Cellular & Molecular Immunology, 2:439-448, 2005; Brinkmann et al, Pharmacology & Therapeutics, 115:84-105, 2007;
  • FTY720 has been found to have therapeutic efficacy for multiple sclerosis in clinical trials. In Phase II clinical trials for relapsing-remitting multiple sclerosis, FTY720 was found to reduce the number of lesions detected by magnetic resonance imaging (MRI) and clinical disease activity in patients with multiple sclerosis (Kappos et al, N. Engl. J. Med., 355:1124-1140, 2006; Martini et al, Expert Opin. Investig.
  • MRI magnetic resonance imaging
  • FTY720 is currently in Phase
  • FTY720 has been reported to impair migration of dendritic cells infected with
  • Francisella tularensis to the mediastinal lymph node, thereby reducing the bacterial
  • Francisella tularensis is associated with tularemia, ulceroglandular infection, respiratory infection and a typhoidal disease (E. Bar-Haim et al, PLoS Pathog 4(11): el000211. doi:10.1371/journal.ppat.l000211, 2008).
  • Agonism of the S 1 P 1 receptor has been implicated in enhancement of survival of oligodendrocyte progenitor cells. Survival of oligodendrocyte progenitor cells is a required component of the remyelination process. Remyelination of multiple sclerosis lesions is considered to promote recovery from clinical relapses. (Miron et al, Ann. Neurol., 63:61-71, 2008; Coelho et al, J. Pharmacol Exp. Ther., 323:626-635, 2007; Dev et al, Pharmacology and Therapeutics, 117:77-93, 2008).
  • S1P1 receptor plays a role in platelet-derived growth factor (PDGF)-induced oligodendrocyte progenitor cell mitogenesis (Jung et al, Glia, 55:1656-1667, 2007).
  • PDGF platelet-derived growth factor
  • Agonism of the SIP 1 receptor has also been reported to mediate migration of neural stem cells toward injured areas of the central nervous system (CNS), including in a rat model of spinal cord injury (Kimura et al, Stem Cells, 25 : 115-124, 2007).
  • Agonism of the SIP 1 receptor has been implicated in the inhibition of keratinocyte proliferation (Sauer et al, J. Biol Chem., 279:38471-38479, 2004), consistent with reports that SIP inhibits keratinocyte proliferation (Kim et al, Cell Signal, 16:89-95, 2004).
  • the hyperproliferation of keratinocytes at the entrance to the hair follicle, which can then become blocked, and an associated inflammation are significant pathogenetic factors of acne (Koreck et al, Dermatology, 206:96-105, 2003; Webster, Cutis, 76:4-7, 2005).
  • FTY720 has been reported to have therapeutic efficacy in inhibiting pathologic angiogenesis, such as that as may occur in tumor development. Inhibition of angiogenesis by
  • FTY720 is thought to involve agonism of the S1P1 receptor (Oo et al, J. Biol. Chem.,
  • FTY720 has been reported to have therapeutic efficacy for inhibiting primary and metastatic tumor growth in a mouse model of melanoma (LaMontagne et al., Cancer Res., 66:221 -231 , 2006). FTY720 has been reported to have therapeutic efficacy in a mouse model for metastatic hepatocellular carcinoma (Lee et al, Clin. Cancer Res., 11:84588466, 2005).
  • Cyclosporin A and FK506 are drugs used to prevent rejection of transplanted organs. Although they are effective in delaying or suppressing transplant rejection, classical immunosuppressants such as cyclosporin A and FK506 are known to cause several undesirable side effects including nephrotoxicity, neurotoxicity, /3-cell toxicity and
  • immunosuppressant without these side effects which is effective as a monotherapy or in combination with a classical immunosuppressant for inhibiting migration of, e.g. , alloantigen- reactive T-cells to the grafted tissue, thereby prolonging graft survival.
  • FTY720 has been shown to have therapeutic efficacy in transplant rejection both as a monotherapy and in synergistic combination with a classical immunosuppressant, including cyclosporin A, FK506 and RAD (an mTOR inhibitor). It has been shown that, unlike the classical immunosuppressants cyclosporin A, FK506 and RAD, FTY720 has efficacy for prolonging graft survival without inducing general immunosuppression, and this difference in drug action is believed to be relevant to the synergism observed for the combination (Brinkmann et al, Transplant Proc, 33:530-531, 2001; Brinkmann et al, Transplantation, 72:764-769, 2001).
  • a classical immunosuppressant including cyclosporin A, FK506 and RAD (an mTOR inhibitor). It has been shown that, unlike the classical immunosuppressants cyclosporin A, FK506 and RAD, FTY720 has efficacy for prolonging graft survival without
  • Agonism of the S1P1 receptor has been reported to have therapeutic efficacy for prolonging allograft survival in mouse and rat skin allograft models (Lima et al, Transplant Proc, 36:1015-1017, 2004; Yan et al, Bioorg. & Med. Chem. Lett., 16:3679-3683, 2006).
  • FTY720 has been reported to have therapeutic efficacy for prolonging allograft survival in a rat cardiac allograft model (Suzuki et al, Transpl Immunol, 4:252-255, 1996). FTY720 has been reported to act synergistically with cyclosporin A to prolong rat skin allograft survival
  • KRP-203 an SIP receptor agonist has been reported to have therapeutic efficacy for prolonging allograft survival in a rat skin allograft model and both as monotherapy and in synergistic combination with cyclosporin A in a rat cardiac allograft model (Shimizu et al, Circulation, 111 :222-229, 2005).
  • KRP-203 also has been reported to have therapeutic efficacy in combination with mycophenolate mofetil (MMF; a prodrug for which the active metabolite is mycophenolic acid, an inhibitor of purine biosynthesis) for prolonging allograft survival both in a rat renal allograft model and in a rat cardiac allograft model (Suzuki et al, J.
  • MMF mycophenolate mofetil
  • FTY720 has been reported to have therapeutic efficacy in a mouse islet graft model (Fu et al, Transplantation, 73:1425-1430, 2002; Liu et al, Microsurgery, 27:300-304; 2007) and in a study using human islet cells to evidence no detrimental effects on human islet function (Truong et al, American Journal of Transplantation, 7:2031-2038, 2007).
  • FTY720 has been reported to reduce the nociceptive behavior in the spared nerve injury model for neuropathic pain which does not depend on prostaglandin synthesis (O. Costu et al, Journal of Cellular and Molecular Medicine 12(3), 995-1004, 2008).
  • FTY720 has been reported to impair initiation of murine contact hypersensitivity (CHS). Adoptive transfer of immunized lymph node cells from mice treated with FTY720 during the sensitization phase was virtually incapable of inducing CHS response in recipients (D. Nakashima et al, J. Investigative Dermatology (128(12), 2833-2841, 2008).
  • the present invention encompasses compounds which are agonists of the SlPl receptor having selectivity over the S1P3 receptor.
  • the S1P3 receptor and not the SlPl receptor, has been directly implicated in bradycardia (Sanna et al, J. Biol. Chem., 279: 13839-13848, 2004).
  • An SlPl receptor agonist selective over at least the S1P3 receptor has advantages over current therapies by virtue of an enhanced therapeutic window, allowing better tolerability with higher dosing and thus improving efficacy as therapy.
  • the present invention encompasses compounds which are agonists of the S1P1 receptor and which exhibit no or substantially no activity for bradycardia.
  • S1P1 receptor agonists are useful for treating or preventing conditions where suppression of the immune system or agonism of the S1P1 receptor is in order, such as diseases and disorders mediated by lymphocytes, transplant rejection, autoimmune diseases and disorders, inflammatory diseases and disorders, and conditions that have an underlying defect in vascular integrity or that relate to angiogenesis such as may be pathologic.
  • S1P1 receptor agonists are useful for treating or preventing conditions where suppression of the immune system or agonism of the S1P1 receptor is in order, such as diseases and disorders mediated by lymphocytes, transplant rejection, autoimmune diseases and disorders, inflammatory diseases and disorders (e.g., acute and chronic inflammatory conditions), cancer, and conditions that have an underlying defect in vascular integrity or that are associated with angiogenesis such as may be pathologic (e.g., as may occur in inflammation, tumor development and atherosclerosis).
  • diseases and disorders mediated by lymphocytes include diseases and disorders mediated by lymphocytes; conditions that have an underlying defect in vascular integrity; autoimmune diseases and disorders; inflammatory diseases and disorders (e.g., acute and chronic
  • arthritis including psoriatic arthritis, and rheumatoid arthritis
  • diabetes including type I diabetes
  • demyelinating disease including multiple sclerosis
  • ischemia-reperfusion injury including renal and cardiac ischemia-reperfusion injury
  • inflammatory skin disease including psoriasis, atopic dermatitis, and acne
  • hyperproliferative skin disease including acne
  • inflammatory bowel disease including Crohn's disease, and ulcerative colitis
  • systemic lupus erythematosis systemic lupus erythematosis
  • asthma uveitis
  • myocarditis allergy
  • atherosclerosis brain inflammation, including Alzheimer's disease, and brain inflammatory reaction following traumatic brain injury
  • central nervous system disease including spinal cord injury, or cerebral infarction
  • pathologic angiogenesis including as may occur in primary and metastatic tumor growth
  • S1P1 receptor agonists are useful for treating microbial infections, and viral infections or diseases.
  • the S1P1 receptor-associated disorder is a disease or disorder mediated by lymphocytes.
  • the S1P1 receptor-associated disorder is an autoimmune disease or disorder. In some embodiments, the S1P1 receptor-associated disorder is an inflammatory disease or disorder.
  • the S1P1 receptor-associated disorder is a microbial infection or microbial disease.
  • the S1P1 receptor-associated disorder is a viral infection or viral disease.
  • the S1P1 receptor-associated disorder is cancer.
  • the S1P1 receptor-associated disorder is a disorder in an individual, wherein the disorder is selected from the group consisting of: psoriasis, rheumatoid arthritis, Crohn's disease, transplant rejection, multiple sclerosis, systemic lupus erythematosus, ulcerative colitis, type I diabetes, acne, myocardial ischemia-reperfusion injury, hypertensive nephropathy, glomerulosclerosis, gastritis, polymyositis, thyroiditis, vitiligo, hepatitis, and biliary cirrhosis.
  • the disorder is selected from the group consisting of: psoriasis, rheumatoid arthritis, Crohn's disease, transplant rejection, multiple sclerosis, systemic lupus erythematosus, ulcerative colitis, type I diabetes, acne, myocardial ischemia-reperfusion injury, hypertensive nephropathy, glomerulos
  • the S1P1 receptor-associated disorder is psoriasis.
  • the S1P1 receptor-associated disorder is rheumatoid arthritis.
  • the S1P1 receptor-associated disorder is Crohn's disease.
  • the S1P1 receptor-associated disorder is transplant rejection.
  • the SI PI receptor-associated disorder is multiple sclerosis.
  • the S1P1 receptor-associated disorder is systemic lupus erythematosus.
  • the S1P1 receptor-associated disorder is ulcerative colitis.
  • the S1P1 receptor-associated disorder is type I diabetes.
  • the S1P1 receptor-associated disorder is acne.
  • the S1P1 receptor-associated disorder is myocardial ischemia- reperfusion injury.
  • the S1P1 receptor-associated disorder is hypertensive nephropathy.
  • the S1P1 receptor-associated disorder is glomerulosclerosis. In some embodiments, the S1P1 receptor-associated disorder is gastritis.
  • the SI PI receptor-associated disorder is polymyositis.
  • the S1P1 receptor-associated disorder is thyroiditis.
  • the S1P1 receptor-associated disorder is vitiligo.
  • the S1P1 receptor-associated disorder is hepatitis.
  • the S1P1 receptor-associated disorder is biliary cirrhosis.
  • One aspect of the present invention pertains to compounds represented by any of the formulae described herein used in the preparation of pharmaceutical compositions.
  • a further aspect of the present invention pertains to pharmaceutical compositions comprising Compound of Formula (la) or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers. Some embodiments pertain to pharmaceutical compositions comprising Compound of Formula (la) or a pharmaceutically acceptable salt and a pharmaceutically acceptable carrier.
  • Some embodiments of the present invention include a method of producing a pharmaceutical composition comprising admixing Compound of Formula (la) or a
  • Formulations may be prepared by any suitable method, typically by uniformly mixing Compound of Formula (la) or a pharmaceutically acceptable salt thereof with liquids or finely divided solid carriers, or both, in the required proportions and then, if necessary, forming the resulting mixture into a desired shape.
  • excipients such as binding agents, fillers, acceptable wetting agents, tabletting lubricants and disintegrants may be used in tablets and capsules for oral
  • Liquid preparations for oral administration may be in the form of solutions, emulsions, aqueous or oily suspensions and syrups.
  • the oral preparations may be in the form of a dry powder that can be reconstituted with water or another suitable liquid vehicle before use. Additional additives such as suspending or emulsifying agents, non-aqueous vehicles (including edible oils), preservatives and flavorings and colorants may be added to the liquid preparations.
  • Parenteral dosage forms may be prepared by dissolving Compound of Formula (la) or a pharmaceutically acceptable salt thereof in a suitable liquid vehicle and filter sterilizing the solution before filling and sealing an appropriate vial or ampule. These are just a few examples of the many appropriate methods well known in the art for preparing dosage forms.
  • Compound of Formula (la) or a pharmaceutically acceptable salt thereof can be formulated into pharmaceutical compositions using techniques well known to those in the art. Suitable pharmaceutically-acceptable carriers, outside those mentioned herein, are known in the art; for example, see Remington, The Science and Practice of Pharmacy, 20 th Edition, 2000, Lippincott Williams & Wilkins, (Editors: Gennaro et at.)
  • Compound of Formula (la) or a pharmaceutically acceptable salt thereof may, in an alternative use, be administered as a raw or pure chemical, it is preferable however to present the active ingredient as a pharmaceutical formulation or composition further comprising a pharmaceutically acceptable carrier.
  • the invention thus further relates to pharmaceutical formulations comprising
  • compositions include those suitable for oral, rectal, nasal, topical
  • Transdermal patches dispense a drug at a controlled rate by presenting the drug for absorption in an efficient manner with a minimum of degradation of the drug.
  • transdermal patches comprise an impermeable backing layer, a single pressure sensitive adhesive and a removable protective layer with a release liner.
  • Compound of Formula (la) or a pharmaceutically acceptable salt thereof, together with a conventional adjuvant, carrier, or diluent, may thus be placed into the form of pharmaceutical formulations and unit dosages thereof and in such form may be employed as solids, such as tablets or filled capsules, or liquids such as solutions, suspensions, emulsions, elixirs, gels or capsules filled with the same, all for oral use, in the form of suppositories for rectal
  • compositions and unit dosage forms thereof may comprise conventional ingredients in conventional proportions, with or without additional active ingredients and such unit dosage forms may contain any suitable effective amount of the active ingredient commensurate with the intended daily dosage range to be employed.
  • the pharmaceutical composition may be in the form of, for example, a tablet, capsule, suspension or liquid.
  • the pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient.
  • Examples of such dosage units are capsules, tablets, powders, granules or suspensions, with conventional additives such as lactose, mannitol, corn starch or potato starch; with binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators such as corn starch, potato starch or sodium carboxymethyl-cellulose; and with lubricants such as talc or magnesium stearate.
  • the active ingredient may also be administered by injection as a composition wherein, for example, saline, dextrose or water may be used as a suitable pharmaceutically acceptable carrier.
  • Compound of Formula (la) and pharmaceutically acceptable salts thereof can be used as active ingredients in pharmaceutical compositions, specifically as S1P1 receptor modulators.
  • active ingredient in the context of a “pharmaceutical composition” is intended to mean a component of a pharmaceutical composition that provides the primary pharmacological effect, as opposed to an "inactive ingredient” which would generally be recognized as providing no pharmaceutical benefit.
  • the dose when using Compound of Formula (la) or a pharmaceutically acceptable salt thereof can vary within wide limits and as is customary and as is known to the physician, it is to be tailored to the individual conditions in each individual case. It depends, for example, on the nature and severity of the illness to be treated; on the condition of the patient; on the formulation employed; on whether an acute or chronic disease state is treated, or prophylaxis is conducted; or on whether further active ingredients are administered in addition to Compound of Formula (la) or a pharmaceutically acceptable salt thereof.
  • Representative doses of the present invention include, but are not limited to, about 0.001 mg to about 5000 mg, about 0.001 mg to about 2500 mg, about 0.001 mg to about 1000 mg, 0.001 mg to about 500 mg, 0.001 mg to about 250 mg, about 0.001 mg to 100 mg, about 0.001 mg to about 50 mg and about 0.001 mg to about 25 mg.
  • Multiple doses may be administered during the day, especially when relatively large amounts are deemed to be needed, for example two, three or four doses. Depending on the individual and as deemed appropriate from the patient's physician or caregiver it may be necessary to deviate upward or downward from the doses described herein.
  • the amount of active ingredient required for use in treatment will vary not only with the particular ingredient selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will ultimately be at the discretion of the attendant physician or clinician.
  • a model system typically an animal model
  • these extrapolations may merely be based on the weight of the animal model in comparison to another, such as a mammal, preferably a human, however, more often, these extrapolations are not simply based on weights, but rather incorporate a variety of factors.
  • compositions include the type, age, weight, sex, diet and medical condition of the patient, the severity of the disease, the route of administration, pharmacological considerations such as the activity, efficacy, pharmacokinetic and toxicology profiles of the particular active ingredient employed, whether a drug delivery system is utilized, on whether an acute or chronic disease state is being treated or prophylaxis is conducted or on whether further active ingredients are administered in addition to Compound of Formula (la) or a pharmaceutically acceptable salt thereof as part of a drug combination.
  • the dosage regimen for treating a disease condition with Compound of Formula (la) or a pharmaceutically acceptable salt thereof is selected in accordance with a variety factors as cited above. Thus, the actual dosage regimen employed may vary widely and therefore may deviate from a preferred dosage regimen and one skilled in the art will recognize that dosages and dosage regimens outside these typical ranges can be tested and, where appropriate, may be used in the methods described herein.
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, for example, into a number of discrete loosely spaced administrations.
  • the daily dose can be divided, especially when relatively large amounts are administered as deemed appropriate, into several, for example, two, three or four part administrations. If appropriate, depending on individual behavior, it may be necessary to deviate upward or downward from the daily dose indicated.
  • Compound of Formula (la) or a pharmaceutically acceptable salt thereof can be administrated in a wide variety of oral and parenteral dosage forms. It will be obvious to those skilled in the art that the following dosage forms may comprise, as the active component, either Compound of Formula (la) or a pharmaceutically acceptable salt thereof.
  • the pharmaceutically acceptable carrier can be either solid, liquid or a mixture of both.
  • Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories and dispersible granules.
  • a solid carrier can be one or more substances which may also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
  • the carrier is a finely divided solid which is in a mixture with the finely divided active component.
  • the active component is mixed with the carrier having the necessary binding capacity in suitable proportions and compacted to the desired shape and size.
  • the powders and tablets may contain varying percentages of the active ingredient.
  • a powder or tablet may contain from 0.5 to about 90 percent of the active ingredient; however, an artisan would know when amounts outside of this range are necessary.
  • Suitable carriers for powders and tablets are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter and the like.
  • the term "preparation" is intended to include the formulation of the active ingredient with encapsulating material as a carrier providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, and which is thus in association with it. Similarly, cachets and lozenges are included.
  • Tablets, powders, capsules, pills, cachets and lozenges can be used as solid forms suitable for oral administration.
  • a low melting wax such as an admixture of fatty acid glycerides or cocoa butter
  • the active component is dispersed homogeneously therein, as by stirring.
  • the molten homogenous mixture is then poured into convenient sized molds, allowed to cool and thereby to solidify.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or sprays containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
  • Liquid form preparations include solutions, suspensions and emulsions, for example, water or water-propylene glycol solutions.
  • parenteral injection liquid preparations can be formulated as solutions in aqueous polyethylene glycol solution.
  • injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • Suitable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Compound of Formula (la) or a pharmaceutically acceptable salt thereof may thus be formulated for parenteral administration (e.g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative.
  • parenteral administration e.g. by injection, for example bolus injection or continuous infusion
  • compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
  • Aqueous formulations suitable for oral use can be prepared by dissolving or suspending the active component in water and adding suitable colorants, flavors, stabilizing and thickening agents, as desired.
  • Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well-known suspending agents.
  • viscous material such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well-known suspending agents.
  • solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for oral administration.
  • liquid forms include solutions, suspensions and emulsions.
  • These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents and the like.
  • pharmaceutically acceptable salt thereof may be formulated as ointments, creams or lotions, or as a transdermal patch.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
  • Formulations suitable for topical administration in the mouth include lozenges comprising active agent in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Solutions or suspensions are applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray.
  • the formulations may be provided in single or multi-dose form. In the latter case of a dropper or pipette, this may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension. In the case of a spray, this may be achieved for example by means of a metering atomizing spray pump.
  • Administration to the respiratory tract may also be achieved by means of an aerosol formulation in which the active ingredient is provided in a pressurized pack with a suitable propellant. If Compound of Formula (la) or a pharmaceutically acceptable salt thereof or pharmaceutical compositions comprising them are administered as aerosols, for example as nasal aerosols or by inhalation, this can be carried out, for example, using a spray, a nebulizer, a pump nebulizer, an inhalation apparatus, a metered inhaler or a dry powder inhaler.
  • compositions for administration of Compound of Formula (la) or a pharmaceutically acceptable salt thereof as an aerosol can be prepared by processes well known to the person skilled in the art.
  • solutions or dispersions of Compound of Formula (la) or a pharmaceutically acceptable salt thereof in water water/alcohol mixtures or suitable saline solutions can be employed using customary additives, for example benzyl alcohol or other suitable preservatives, absorption enhancers for increasing the bioavailability, solubilizers, dispersants and others and, if appropriate, customary propellants, for example include carbon dioxide, CFCs, such as, dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane; and the like.
  • the aerosol may conveniently also contain a surfactant such as lecithin.
  • the dose of drug may be controlled by provision of a metered valve.
  • the pharmaceutical composition will generally have a small particle size for example of the order of 10 microns or less. Such a particle size may be obtained by means known in the art, for example by micronization.
  • formulations adapted to give sustained release of the active ingredient may be employed.
  • the active ingredients may be provided in the form of a dry powder, for example, a powder mix of the salt in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • a powder mix of the salt in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • the powder carrier will form a gel in the nasal cavity.
  • the powder composition may be presented in unit dose form for example in capsules or cartridges of, e.g., gelatin, or blister packs from which the powder may be administered by means of an inhaler.
  • the pharmaceutical preparations are preferably in unit dosage forms.
  • the preparation is subdivided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules and powders in vials or ampoules.
  • the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
  • Tablets or capsules for oral administration and liquids for intravenous administration are preferred compositions.
  • Pharmaceutically acceptable salts including pharmaceutically acceptable acid addition salts may be prepared from pharmaceutically acceptable non-toxic acids including inorganic and organic acids.
  • Representative acids include, but are not limited to, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethenesulfonic, dichloroacetic, formic, fumaric, gluconic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, oxalic, pamoic, pantothenic, phosphoric, succinic, sulfiric, tartaric, oxalic, p-toluenesulfonic and the like, such as those pharmaceutically acceptable salts listed in Berge, et al, Journal of Pharmaceutical Sciences, 66:1-19 (1977).
  • the acid addition salts may be obtained as the direct products of compound synthesis.
  • the free base may be dissolved in a suitable solvent containing the appropriate acid and the salt isolated by evaporating the solvent or otherwise separating the salt and solvent.
  • Compound of Formula (la) or a pharmaceutically acceptable salt thereof may form solvates with standard low molecular weight solvents using methods known to the skilled artisan.
  • Pro-drugs refers to compounds that have been modified with specific chemical groups known in the art and when administered into an individual these groups undergo biotransformation to give the parent compound. Pro-drugs can thus be viewed as compounds containing one or more specialized non-toxic protective groups used in a transient manner to alter or to eliminate a property of the compound. In one general aspect, the "prodrug” approach is utilized to facilitate oral absorption.
  • Some embodiments of the present invention include a method of producing a pharmaceutical composition for "combination-therapy” comprising admixing Compound of Formula (la) or a pharmaceutically acceptable salt thereof, together with at least one known pharmaceutical agent as described herein and a pharmaceutically acceptable carrier.
  • compositions these are not intended for use only in humans, but in other non- human mammals as well. Indeed, recent advances in the area of animal health-care mandate that consideration be given for the use of active agents, such as SIP 1 modulators, for the treatment of an S1P1 -associated disease or disorder in companionship animals (e.g., cats, dogs, etc.) and in livestock animals (e.g., cows, chickens, fish, etc.) Those of ordinary skill in the art are readily credited with understanding the utility of such salts in such settings.
  • active agents such as SIP 1 modulators
  • Compound of Formula (la) and pharmaceutically acceptable salts, solvates and hydrates thereof can be administrated in a wide variety of oral and parenteral dosage forms. It will be apparent to those skilled in the art that the dosage forms may comprise, as the active component, either Compound of Formula (la) and pharmaceutically acceptable salts, solvates and hydrates thereof. Moreover, various hydrates and solvates of Compound of Formula (la) and
  • one aspect of the present invention relates to processes for preparing hydrates and solvates of Compound of Formula (la) and/or pharmaceutically acceptable salts thereof, as described herein, that can be isolated and characterized by methods known in the art, such as, thermogravimetric analysis (TGA), TGA- mass spectroscopy, TGA-Infrared spectroscopy, powder X-ray diffraction (PXRD), Karl Fisher titration, high resolution X-ray diffraction, and the like.
  • TGA thermogravimetric analysis
  • TGA- mass spectroscopy TGA- mass spectroscopy
  • TGA-Infrared spectroscopy powder X-ray diffraction (PXRD)
  • Karl Fisher titration high resolution X-ray diffraction
  • Example companies offering these services include Wilmington PharmaTech (Wilmington, DE),
  • LCMS spec HPLC-pumps: LC-IOAD VP, Shimadzu Inc.; HPLC system controller: SCL-IOA VP, Shimadzu Inc; UV-Detector: SPD-10A VP, Shimadzu Inc; Autosampler: CTC HTS, PAL, Leap Scientific; Mass spectrometer: API 150EX with Turbo Ion Spray source, AB/MDS Sciex; Software: Analyst 1.2.
  • the reactor content was allowed to cool below 20 °C and a mixture of l-bromo-2-(trifluoromethyl)benzene (1500 g, 6.67 mol) and bromocyclopentane (1192 g, 8.00 mol, 1.2 eq.) added dropwise under N 2 at such a rate as to maintain the internal temperature between 25-30 °C (caution, exothermic). After the addition (3.33 h) and the exotherm subsided, the reaction mixture was stirred at 25 °C under N 2 overnight, allowed to cool to an internal temperature of 0 °C and quenched with 6 N HCL (3 L, 1.5 h) at such a rate as to maintain the internal temperature below 15 °C (caution, very exothermic).
  • Step A Preparation of l-(2-(Trifluoromethyl)phenyl)cyclopentanol.
  • the reaction mixture was stirred at -78 °C for 30 min, gradually brought to room temperature, and stirred for 1 h.
  • the reaction mixture was cooled by an ice bath, quenched with water, and acidified to pH 4-5 by addition of concentrated HC1.
  • the solvent was removed under reduced pressure.
  • the residue was dissolved in methylene chloride, washed with water (2 times), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure.
  • the residue was purified by silica gel chromatography to give the title compound as an oil (250 mg).
  • LCMS m/z 213.1 [M-H 2 0+H] + .
  • Step B Preparation of l-Cyclopentyl-2-(trifluoromethyl)benzene.
  • Step A Preparation of Methyl 4-Chloro-3-(trifiuoromethyl)benzoate.
  • Step B Preparation of Methyl 4-Cyclopentyl-3-(trifluoromethyl)benzoate.
  • Step C Preparation of (4-Cyclopentyl-3-(trifluoromethyl)phenyl)methanol.
  • Step D Preparation of 4-(Chloromethyl)-l-cyclopentyl-2-(trifluoromethyl)benzene.
  • Step A Preparation of Methyl 4-Chloro-3-(trifluoromethyl)benzoate.
  • Step B Preparation of Methyl 4-Cyclopentyl-3-(trifluoromethyl)benzoate.
  • Step C Preparation of (4-Cyclopentyl-3-(trifluoromethyl)phenyl)methanol.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Diabetes (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Rheumatology (AREA)
  • Urology & Nephrology (AREA)
  • Dermatology (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Neurosurgery (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Vascular Medicine (AREA)
  • Obesity (AREA)
  • Pain & Pain Management (AREA)
  • Emergency Medicine (AREA)
EP11703939A 2010-01-27 2011-01-27 Verfahren zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen daraus Withdrawn EP2528894A1 (de)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP22193921.8A EP4148045A1 (de) 2010-01-27 2011-01-27 Zwischenverbindungen zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen davon
EP18156371.9A EP3378854B1 (de) 2010-01-27 2011-01-27 Verfahren zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen davon

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US33683510P 2010-01-27 2010-01-27
PCT/US2011/000153 WO2011094008A1 (en) 2010-01-27 2011-01-27 Processes for the preparation of (r)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP22193921.8A Division EP4148045A1 (de) 2010-01-27 2011-01-27 Zwischenverbindungen zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen davon
EP18156371.9A Division EP3378854B1 (de) 2010-01-27 2011-01-27 Verfahren zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen davon

Publications (1)

Publication Number Publication Date
EP2528894A1 true EP2528894A1 (de) 2012-12-05

Family

ID=43648279

Family Applications (3)

Application Number Title Priority Date Filing Date
EP22193921.8A Pending EP4148045A1 (de) 2010-01-27 2011-01-27 Zwischenverbindungen zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen davon
EP18156371.9A Active EP3378854B1 (de) 2010-01-27 2011-01-27 Verfahren zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen davon
EP11703939A Withdrawn EP2528894A1 (de) 2010-01-27 2011-01-27 Verfahren zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen daraus

Family Applications Before (2)

Application Number Title Priority Date Filing Date
EP22193921.8A Pending EP4148045A1 (de) 2010-01-27 2011-01-27 Zwischenverbindungen zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen davon
EP18156371.9A Active EP3378854B1 (de) 2010-01-27 2011-01-27 Verfahren zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen davon

Country Status (8)

Country Link
US (5) US8853419B2 (de)
EP (3) EP4148045A1 (de)
JP (2) JP5856980B2 (de)
CN (2) CN103221391B (de)
CA (1) CA2786994C (de)
ES (1) ES2937386T3 (de)
SG (3) SG10201906876PA (de)
WO (1) WO2011094008A1 (de)

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20180023049A (ko) 2008-07-23 2018-03-06 아레나 파마슈티칼스, 인크. 자가면역성 및 염증성의 장애의 치료에 유용한 치환된 1,2,3,4-테트라히드로시클로펜타[b]인돌-3-일)아세트산 유도체
EP2342205B1 (de) 2008-08-27 2016-04-20 Arena Pharmaceuticals, Inc. Substituierte tricyclische carbonsäurederivative als s1p1 rezeptor agonisten für die behandlung von entzündlichen und autoimmunerkrankungen
EP4148045A1 (de) * 2010-01-27 2023-03-15 Arena Pharmaceuticals, Inc. Zwischenverbindungen zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen davon
SG10201501575VA (en) 2010-03-03 2015-04-29 Arena Pharm Inc Processes for the preparation of s1p1 receptor modulators and crystalline forms thereof
CN102659664B (zh) * 2012-03-28 2015-01-21 中国计量学院 合成分离拉洛皮兰及其类似物的方法
JP6895378B2 (ja) * 2015-01-06 2021-06-30 アリーナ ファーマシューティカルズ, インコーポレイテッド S1p1受容体に関連する状態の処置方法
EP3939965A1 (de) * 2015-06-22 2022-01-19 Arena Pharmaceuticals, Inc. Kristallines l-arginin-salz von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta[b]indol-3-yl)essigsäure zur verwendung in s1p1-rezeptor-assoziierten erkrankungen
CN106349006B (zh) * 2016-08-26 2019-02-05 大连奇凯医药科技有限公司 3-三氟甲基苯乙腈的制备方法
CN110520124A (zh) * 2017-02-16 2019-11-29 艾尼纳制药公司 用于治疗原发性胆汁性胆管炎的化合物和方法
WO2018151834A1 (en) 2017-02-16 2018-08-23 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of inflammatory bowel disease with extra-intestinal manifestations
US11066422B2 (en) 2017-07-13 2021-07-20 Toray Fine Chemicals Co., Ltd. Method of producing cycloalkyl(trifluoromethyl)benzene
CA3102136A1 (en) 2018-06-06 2019-12-12 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the s1p1 receptor
KR20210074291A (ko) 2018-09-06 2021-06-21 아레나 파마슈티칼스, 인크. 자가면역 및 염증성 장애의 치료에 유용한 화합물
WO2020072824A1 (en) 2018-10-03 2020-04-09 Arena Pharmaceuticals, Inc. Methods for the treatment of scleroderma
CN109529920B (zh) * 2018-11-12 2021-11-02 山东科技大学 用于轻质烷烃异构化的负载硼化钛催化剂及其制备方法和使用方法
CN109678715B (zh) * 2018-11-28 2019-11-12 中国药科大学 2-(1-酰氧正戊基)苯甲酸与碱性氨基酸或氨基胍形成的盐、其制备方法及用途
CN113226307A (zh) 2018-11-30 2021-08-06 艾尼纳制药公司 治疗与s1p1受体有关的病况的方法
EP3908276A1 (de) 2019-01-08 2021-11-17 Arena Pharmaceuticals, Inc. Verfahren zur behandlung von zuständen im zusammenhang mit dem s1p1-rezeptor
MX2022003982A (es) * 2019-10-01 2022-07-12 Arena Pharm Inc Metodos de tratamiento de enfermedades relacionadas con el receptor s1p1.
AU2020372647A1 (en) 2019-10-31 2022-06-16 Idorsia Pharmaceuticals Ltd Combination of a CXCR7 antagonist with an S1P1 receptor modulator
CN111763148B (zh) * 2020-06-19 2022-10-28 广州大学 一种含三氟甲基的炔基环戊烯衍生物及其制备方法和应用
CN114105745A (zh) * 2020-08-28 2022-03-01 广东东阳光药业有限公司 辛波莫德中间体及其制备方法
CN117597119A (zh) * 2021-07-02 2024-02-23 阿芮纳制药有限公司 向患有肝损伤的个体施用化合物
EP4212156A1 (de) 2022-01-13 2023-07-19 Abivax Kombination von 8-chlor-n-(4-trifluormethoxy)phenyl)chinolin-2-amin und dessen derivaten mit einem s1p-rezeptormodulator
WO2023135506A1 (en) 2022-01-13 2023-07-20 Arena Pharmaceuticals, Inc. Etrasimod for use in treating s1p1 receptor-associated disorders in combination with hormone treatment
WO2023214312A1 (en) 2022-05-06 2023-11-09 Arena Pharmaceuticals, Inc. Methods of treating atopic dermatitis with etrasimod

Family Cites Families (272)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL293572A (de) 1962-06-07
CH478818A (de) 1965-10-22 1969-09-30 Ciba Geigy Verfahren zur Herstellung neuer N,N'-Di-(pyrimidyl-(4)-aminoalkyl)-diazacycloalkanen
CH480410A (de) 1967-01-09 1969-10-31 Geigy Ag J R Verfahren zur Herstellung von wasserlöslichen Azopyrimidinfarbstoffen
CA961052A (en) 1967-01-12 1975-01-14 Max Schellenbaum N-2-ethylhexyl-n'-aryl ureas and preparation containing them
US3608087A (en) 1968-06-21 1971-09-21 Merck & Co Inc Feed compositions
US3887329A (en) 1969-05-05 1975-06-03 Ciba Geigy Ag Hexamethyl phosphotriamide-dye compositions
US3686238A (en) 1970-01-19 1972-08-22 Syntex Corp Glycerol esterified with 2-naphthyl-acetic acids and fatty acids
US3852434A (en) 1970-09-11 1974-12-03 Merck & Co Inc Potentiation of ({31 ) cis-1,2-epoxypropyl)phosphonic acid and analogues thereof
US3690834A (en) 1971-01-11 1972-09-12 Syva Co Ligand determination with spin labeled compounds by receptor displacement
US3966744A (en) 1971-01-11 1976-06-29 Syva Company Spin labeled compounds
DE2106585A1 (de) 1971-02-11 1972-08-24 Farbenfabriken Bayer Ag, 5090 Leverkusen Aminothiodiazole und Thiodiazol-Azofarbstoffe
DE2226703A1 (de) 1972-05-25 1973-12-13 Schering Ag Neue tetrahydrocarbazolderivate und verfahren zu ihrer herstellung
US3966764A (en) 1972-07-10 1976-06-29 Syva Company Ligand determination of spin labeled compounds by receptor displacement-amphetamine analogs
US3849420A (en) 1972-10-20 1974-11-19 Dow Chemical Co Bis-(alkylthio-and alkylsulfonyl)-pentachloroquinolines
CH574206A5 (de) 1972-11-16 1976-04-15 Ciba Geigy Ag
DE2340569C2 (de) 1973-08-10 1982-12-02 Bayer Ag, 5090 Leverkusen Azofarbstoffe
AT340933B (de) 1973-08-20 1978-01-10 Thomae Gmbh Dr K Verfahren zur herstellung neuer pyrimidinderivate und ihrer saureadditionssalze
US4057559A (en) 1973-10-01 1977-11-08 American Home Products Corporation Carbazole acetic acid derivatives
US4101541A (en) 1973-12-21 1978-07-18 Ciba-Geigy Corporation 3-Cyano-1,2,4-thiadiazolyl-5-czo dyestuffs
FR2306697A1 (fr) 1975-04-10 1976-11-05 Sogeras Nouvelles pyrimidines utilisables comme medicaments antidiabetiques et hypocholesterolemiants
US4139705A (en) 1977-05-20 1979-02-13 The Dow Chemical Company Pyrazolopyrimidines
US4189579A (en) 1977-05-20 1980-02-19 The Dow Chemical Company Aminoalkylthiopurines
DE2731264A1 (de) 1977-07-11 1979-02-01 Boehringer Mannheim Gmbh Neue 1-acyl-2-cyanaziridine, verfahren zu deren herstellung und diese verbindungen enthaltende pharmazeutische zubereitungen
JPS6038696B2 (ja) 1977-12-09 1985-09-02 コニカ株式会社 ハロゲン化銀カラ−写真感光材料
US4242507A (en) 1978-02-23 1980-12-30 Fujisawa Pharmaceutical Co., Ltd. Sulfonic acid esters
DE2831580C2 (de) 1978-07-18 1980-09-18 Boehringer Mannheim Gmbh, 6800 Mannheim Verfahren und Reagens zur Bestimmung von Glycerin
DE2906603A1 (de) 1979-02-21 1980-09-04 Boehringer Mannheim Gmbh N-substituierte aziridin-2-carbonsaeurederivate, verfahren zu deren herstellung sowie diese substanzen enthaltende arzneimittel
US4343804A (en) 1979-03-26 1982-08-10 A. H. Robins Company, Inc. 4-Amino-3-quinolinecarboxylic acids and esters-antisecretory anti-ulcer compounds
DOP1981004033A (es) 1980-12-23 1990-12-29 Ciba Geigy Ag Procedimiento para proteger plantas de cultivo de la accion fitotoxica de herbicidas.
US4476248A (en) 1983-02-28 1984-10-09 The Upjohn Company Crystallization of ibuprofen
DE3334455A1 (de) 1983-03-04 1984-09-06 Bayer Ag, 5090 Leverkusen Guanidin - derivate
US4612376A (en) 1983-03-25 1986-09-16 Fujisawa Pharmaceutical Co., Ltd. Substituted-3,4-dihydro-4-(2,4,6-trimethoxyphenylimino)-2(1H)-pyrimidones useful as cardiotonic, antihypertensive, cerebrovascular vasodilator and anti-platelet agent
ZA848275B (en) 1983-12-28 1985-08-28 Degussa New piridine-2-ethers or pyridine-2-thioethers having a nitrogen-containing cycloaliphatic ring
DE3601196A1 (de) 1986-01-17 1987-07-23 Merck Patent Gmbh 1,4-dihydropyridine
US4810699A (en) 1987-02-20 1989-03-07 American Home Products Corporation Substituted 1,3,4,9-tetrahydropyrano[3,4,-b]indole-1-acetic acids, pharmaceutical compositions containing them, and methods for treating inflammatory conditions and for analgesic purposes using them
US4782076A (en) 1988-03-01 1988-11-01 American Home Products Corporation Substituted 2,3,4,9-tetrahydro-1H-carbazole-1-acetic acid derivatives, composition and use
PT95692A (pt) 1989-10-27 1991-09-13 American Home Prod Processo para a preparacao de derivados de acidos indole-,indeno-,piranoindole- e tetra-hidrocarbazole-alcanoicos, ou quais sao uteis como inibidores de pla2 e da lipoxigenase
US5221678A (en) 1990-07-26 1993-06-22 Merck Frosst Canada, Inc. (quinolin-2-ylmethoxy)tetrahydrocarbazoles as inhibitors of the biosynthesis of leukotrienes
DE4208254A1 (de) 1992-03-14 1993-09-16 Hoechst Ag Substituierte pyrimidine, verfahren zu ihrer herstellung und ihre verwendung als schaedlingsbekaempfungsmittel und fungizid
FR2692575B1 (fr) 1992-06-23 1995-06-30 Sanofi Elf Nouveaux derives du pyrazole, procede pour leur preparation et compositions pharmaceutiques les contenant.
US5998499A (en) 1994-03-25 1999-12-07 Dentsply G.M.B.H. Liquid crystalline (meth)acrylate compounds, composition and method
KR960704855A (ko) 1993-10-12 1996-10-09 돈 엠. 커. 1n-알킬-n-아릴피리미딘아민 및 이들의 유도체 (1n-alkyl-n-arylpyrimidinamines and derivatives thereof)
TW530047B (en) 1994-06-08 2003-05-01 Pfizer Corticotropin releasing factor antagonists
CN1118454C (zh) 1994-09-09 2003-08-20 日本新药株式会社 杂环衍生物和医药
US5691364A (en) 1995-03-10 1997-11-25 Berlex Laboratories, Inc. Benzamidine derivatives and their use as anti-coagulants
US6403599B1 (en) 1995-11-08 2002-06-11 Pfizer Inc Corticotropin releasing factor antagonists
US6956047B1 (en) 1995-06-06 2005-10-18 Pfizer Inc. Corticotropin releasing factor antagonists
KR0169813B1 (ko) 1995-07-12 1999-01-15 김종인 4-아미노-3-아실나프티리딘 유도체
US5849759A (en) 1995-12-08 1998-12-15 Berlex Laboratories, Inc. Naphthyl-substituted benzimidazole derivatives as anti-coagulants
US5948786A (en) 1996-04-12 1999-09-07 Sumitomo Pharmaceuticals Company, Limited Piperidinylpyrimidine derivatives
US6060478A (en) 1996-07-24 2000-05-09 Dupont Pharmaceuticals Azolo triazines and pyrimidines
US5776967A (en) 1996-07-26 1998-07-07 American Home Products Corporation Pyranoindole inhibitors of COX--2
AR008789A1 (es) 1996-07-31 2000-02-23 Bayer Corp Piridinas y bifenilos substituidos
US5830911A (en) 1996-08-14 1998-11-03 American Home Products Corporation Pyranoindole and tetrahydrocarbazole inhibitors of COX-2
US6008234A (en) 1996-09-12 1999-12-28 Berlex Laboratories, Inc. Benzamidine derivatives substituted by cyclic amino acid and cyclic hydroxy acid derivatives and their use as anti-coagulants
NL1010018C2 (nl) 1997-09-09 1999-03-10 Duphar Int Res Chinoline en chinazoline derivaten met corticotropine releasing factor (CRF) antagonistische werking.
US6861448B2 (en) 1998-01-14 2005-03-01 Virtual Drug Development, Inc. NAD synthetase inhibitors and uses thereof
US6187777B1 (en) 1998-02-06 2001-02-13 Amgen Inc. Compounds and methods which modulate feeding behavior and related diseases
CA2339188A1 (en) 1998-08-21 2000-03-02 Dupont Pharmaceuticals Company Isoxazolo¬4,5-d|pyrimidines as crf antagonists
US6239126B1 (en) 1998-12-17 2001-05-29 American Home Products Corporation Arylpiperidine and aryl-1,2,5,6-tetra-hydropyridine urea derivatives
CZ27399A3 (cs) 1999-01-26 2000-08-16 Ústav Experimentální Botaniky Av Čr Substituované dusíkaté heterocyklické deriváty, způsob jejich přípravy, tyto deriváty pro použití jako léčiva, farmaceutická kompozice a kombinovaný farmaceutický přípravek tyto deriváty obsahující a použití těchto derivátů pro výrobu léčiv
US6267985B1 (en) 1999-06-30 2001-07-31 Lipocine Inc. Clear oil-containing pharmaceutical compositions
DK1177187T3 (da) 1999-04-28 2007-10-15 Sanofi Aventis Deutschland Diarylsyrederivater som PPAR-receptorligander
WO2001003739A1 (fr) 1999-07-12 2001-01-18 Ono Pharmaceutical Co., Ltd. Inhibiteurs de fibrose contenant comme ingredient actif l'agoniste du recepteur de sphingosine-1-phosphate ou la sphingosine-1-phosphate
PL354249A1 (en) 1999-09-17 2003-12-29 Abbott Gmbhabbott Gmbh Pyrazolopyrimidines as therapeutic agents
US6921763B2 (en) 1999-09-17 2005-07-26 Abbott Laboratories Pyrazolopyrimidines as therapeutic agents
US6414002B1 (en) 1999-09-22 2002-07-02 Bristol-Myers Squibb Company Substituted acid derivatives useful as antidiabetic and antiobesity agents and method
JP2003510327A (ja) 1999-09-30 2003-03-18 ニューロジェン・コーポレーション 特定のアルキレンジアミンで置換された複素環
US6569879B2 (en) 2000-02-18 2003-05-27 Merck & Co., Inc. Aryloxyacetic acids for diabetes and lipid disorders
US6620821B2 (en) 2000-06-15 2003-09-16 Bristol-Myers Squibb Company HMG-CoA reductase inhibitors and method
HUP0301802A3 (en) 2000-06-28 2009-04-28 Teva Pharma Process for producing carvedilol, crystalline carvedilol, process for producing it and pharmaceutical composition containing it
CN1705649A (zh) 2000-06-29 2005-12-07 艾伯特公司 芳基苯基杂环基硫醚衍生物及其作为抑制细胞粘附的抗炎和免疫抑制剂的用途
US6410583B1 (en) 2000-07-25 2002-06-25 Merck Frosst Canada & Co. Cyclopentanoindoles, compositions containing such compounds and methods of treatment
US20030224058A1 (en) 2002-05-24 2003-12-04 Elan Pharma International, Ltd. Nanoparticulate fibrate formulations
US7276249B2 (en) 2002-05-24 2007-10-02 Elan Pharma International, Ltd. Nanoparticulate fibrate formulations
EP1347755A2 (de) 2000-10-31 2003-10-01 Merck & Co., Inc. Benzopyrancarbonsäuren-derivate zur behandlung von diabetes und fettstörungen
US20020058026A1 (en) 2000-11-13 2002-05-16 Milton Hammerly HMG CoA reductase inhibitor medications combined wih CoEnzyme Q-10
WO2002039987A2 (en) 2000-11-14 2002-05-23 Neurosearch A/S Use of malaria parasite anion channel blockers for treating malaria
US20020137755A1 (en) 2000-12-04 2002-09-26 Bilodeau Mark T. Tyrosine kinase inhibitors
US6545016B1 (en) 2000-12-08 2003-04-08 3M Innovative Properties Company Amide substituted imidazopyridines
US6545017B1 (en) 2000-12-08 2003-04-08 3M Innovative Properties Company Urea substituted imidazopyridines
US6525064B1 (en) 2000-12-08 2003-02-25 3M Innovative Properties Company Sulfonamido substituted imidazopyridines
ME00145B (me) 2001-01-26 2010-10-10 Merck Sharp & Dohme Upotreba supstituisanih jedinjenja azetidinona za lečenje sitosterolemije
EP1383778B1 (de) 2001-01-30 2009-10-21 University Of Virginia Patent Foundation Agonisten und antagonisten von sphingosin-1-phosphatrezeptoren
KR100876047B1 (ko) 2001-03-29 2008-12-26 오사까 가스 가부시키가이샤 광활성 화합물 및 감광성 수지 조성물
WO2002092068A1 (fr) 2001-05-10 2002-11-21 Ono Pharmaceutical Co., Ltd. Derives d'acide carboxylique et medicaments contenant ces derives comme principe actif
US6960692B2 (en) 2001-09-27 2005-11-01 Kyorin Pharmaceutical Co., Ltd. Diaryl sulfide derivative, addition salt thereof, and immunosuppressant
US20070225351A1 (en) 2002-01-11 2007-09-27 Lippa Arnold S Methods and compositions for controlling body weight and appetite
CA2472715A1 (en) 2002-01-18 2003-07-31 Merck & Co., Inc. Edg receptor agonists
AU2003205630A1 (en) 2002-01-18 2003-07-30 The Genetics Company Inc. Beta-secretase inhibitors
AU2003216054B2 (en) 2002-01-18 2007-01-04 Merck & Co., Inc. Selective S1P1/Edg1 receptor agonists
EP1482896A4 (de) 2002-03-01 2005-11-23 Merck & Co Inc Aminoalkylphosphonate und verwandte verbindungen als edg-rezeptor-agonisten
AU2003217764A1 (en) 2002-03-01 2003-09-16 Merck & Co., Inc. Aminoalkylphosphonates and related compounds as edg receptor agonists
DE10219435A1 (de) 2002-05-02 2003-11-13 Bayer Cropscience Ag Substituierte Pyrazolo-pyrimidin-4-one
US7057046B2 (en) 2002-05-20 2006-06-06 Bristol-Myers Squibb Company Lactam glycogen phosphorylase inhibitors and method of use
JP4533134B2 (ja) 2002-06-10 2010-09-01 エラン ファーマ インターナショナル,リミティド ナノ粒子ポリコサノール製剤および新規なポリコサノールの組合せ
EP1549640A4 (de) 2002-06-17 2008-08-06 Merck & Co Inc 1-((5-aryl-1,2,4-oxadiazol-3-yl)benzyl)azetidin-3-carboxylate und 1-((5-aryl-1,2,4-oxadiazol-3-yl)benzyl)pyrrolidin-3-carboxylate als edg-rezeptoragonisten
WO2004007472A1 (ja) 2002-07-10 2004-01-22 Ono Pharmaceutical Co., Ltd. Ccr4アンタゴニストおよびその医薬用途
EP1546110A4 (de) 2002-07-30 2008-03-26 Univ Virginia Verbindungen mit wirkung bei der signalübertragung mittels sphingosin-1-phosphat
DE10237722A1 (de) 2002-08-17 2004-08-19 Aventis Pharma Deutschland Gmbh Indol- oder Benzimidazolderivate zur Modulation der IKappaB-Kinase
CA2505322A1 (en) 2002-11-08 2004-05-21 Takeda Pharmaceutical Company Limited Receptor function regulator
US7098235B2 (en) 2002-11-14 2006-08-29 Bristol-Myers Squibb Co. Triglyceride and triglyceride-like prodrugs of glycogen phosphorylase inhibiting compounds
US20040110241A1 (en) 2002-12-06 2004-06-10 Segal Mark S. Materials and methods for monitoring vascular endothelial function
US7220734B2 (en) 2002-12-20 2007-05-22 Merck & Co., Inc. 1-(amino)indanes and (1,2-dihydro-3-amino)-benzofurans, benzothiophenes and indoles as Edg receptor agonists
JO2397B1 (en) 2002-12-20 2007-06-17 ميرك شارب اند دوم كوربوريشن Terazol derivatives as beta-hydroxy steroid dihydrogenase-1 inhibitors
MXPA05007485A (es) 2003-01-14 2006-01-30 Arena Pharm Inc Derivados de arilo y heteroarilo 1,2,3-trisubstituidos como moduladores del metabolismo y la profilaxis y tratamiento de trastornos relacionados con ello tales como diabetes e hiperglicemia.
GB0301259D0 (en) 2003-01-20 2003-02-19 Novartis Ag Organic compounds
EP1594508B1 (de) 2003-02-11 2012-08-08 Irm Llc Neue bicyclische verbindungen und zusammensetzungen
CN100344638C (zh) 2003-02-18 2007-10-24 杏林制药株式会社 氨基膦酸衍生物、其加成盐以及s1p受体调节剂
CA2515963A1 (en) 2003-02-24 2004-09-10 Arena Pharmaceuticals, Inc. Phenyl- and pyridylpiperidine-derivatives as modulators of glucose metabolism
ATE547395T1 (de) 2003-04-30 2012-03-15 Novartis Pharma Gmbh Aminopropanol-derivate als modulatoren des sphingosin-1-phosphat-rezeptors
US7825260B2 (en) 2003-04-30 2010-11-02 Novartis Ag Aminopropanol derivatives as sphingosine-1-phosphate receptor modulators
EP1628970A2 (de) 2003-04-30 2006-03-01 The Institutes of Pharmaceutical Discovery, LLC Heterocyklisch substituierte carbonsäuren als hemmer der protein-tyrosinphosphatase-1b
US7083933B1 (en) 2003-05-09 2006-08-01 Prosidion Limited Methods for identification of modulators of OSGPR116 activity
EP1625123A4 (de) 2003-05-15 2007-08-29 Merck & Co Inc 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazole als s1p-rezeptoragonisten
WO2004104205A2 (en) * 2003-05-16 2004-12-02 Merck & Co., Inc. Enzymatic preparation of chiral indole esters
SI1638551T1 (sl) 2003-05-19 2012-04-30 Irm Llc Imunosupresivne spojine in sestavki
CL2004001120A1 (es) 2003-05-19 2005-04-15 Irm Llc Compuestos derivados de amina sustituidas con heterociclos, inmunosupresores; composicion farmaceutica; y uso para tratar enfermedades mediadas por interacciones de linfocito, tales como enfermedades autoinmunes, inflamatorias, infecciosas, cancer.
EP1644367B1 (de) 2003-05-19 2015-10-14 Novartis AG Die immunreaktion unterdrückende verbindungen und zusammensetzungen
MY150088A (en) 2003-05-19 2013-11-29 Irm Llc Immunosuppressant compounds and compositions
GB0313612D0 (en) 2003-06-12 2003-07-16 Novartis Ag Organic compounds
AR045047A1 (es) 2003-07-11 2005-10-12 Arena Pharm Inc Derivados arilo y heteroarilo trisustituidos como moduladores del metabolismo y de la profilaxis y tratamiento de desordenes relacionados con los mismos
AR045697A1 (es) 2003-07-14 2005-11-09 Arena Pharm Inc Aril y heteroaril derivados fusionados como moduladores del metabolismo y la prevencion y tratamiento de trastornos relacionados con el mismo
US20100267778A1 (en) 2003-08-04 2010-10-21 Shinya Kusuda Diphenyl ether compound, process for producing the same, and use
CA2535704C (en) 2003-08-28 2012-12-11 Novartis Ag Aminopropanol derivatives
CN101407471A (zh) 2003-08-29 2009-04-15 小野药品工业株式会社 能够结合s1p受体的化合物及其药物用途
WO2005023771A1 (ja) 2003-09-05 2005-03-17 Ono Pharmaceutical Co., Ltd. ケモカインレセプターアンタゴニストおよびその医薬用途
CA2539438A1 (en) 2003-10-01 2005-04-14 Merck And Co., Inc. 3,5-aryl, heteroaryl or cycloalkyl substituted-1,2,4-oxadiazoles as s1p receptor agonists
WO2005041899A2 (en) 2003-11-03 2005-05-12 University Of Virginia Patent Foundation Orally available sphingosine 1-phosphate receptor agonists and antagonists
WO2005044780A1 (ja) 2003-11-10 2005-05-19 Kyorin Pharmaceutical Co., Ltd. アミノカルボン酸誘導体とその付加塩及びs1p受容体調節剤
AU2004299456B2 (en) 2003-12-17 2010-10-07 Merck Sharp & Dohme Corp. (3,4-disubstituted)propanoic carboxylates as S1P (Edg) receptor agonists
JP4766384B2 (ja) 2003-12-19 2011-09-07 小野薬品工業株式会社 リゾホスファチジン酸受容体拮抗作用を有する化合物およびその用途
GB0329498D0 (en) 2003-12-19 2004-01-28 Novartis Ag Organic compounds
GB0401332D0 (en) 2004-01-21 2004-02-25 Novartis Ag Organic compounds
JP2007522233A (ja) 2004-02-11 2007-08-09 アムジエン・インコーポレーテツド バニロイド受容体リガンド及び治療におけるそれらの使用
JP2005272453A (ja) 2004-02-24 2005-10-06 Sankyo Co Ltd アミノアルコール化合物
JP4740884B2 (ja) 2004-02-24 2011-08-03 アイアールエム・リミテッド・ライアビリティ・カンパニー 免疫抑制性化合物および組成物
TW200538433A (en) 2004-02-24 2005-12-01 Irm Llc Immunosuppressant compounds and compositiions
CN1934094A (zh) 2004-03-05 2007-03-21 万有制药株式会社 二芳基取代杂环5元环衍生物
GB0405289D0 (en) 2004-03-09 2004-04-21 Novartis Ag Organic compounds
CN1942440B (zh) 2004-04-02 2011-09-14 默沙东公司 用于制备环烷基并吲哚衍生物的不对称氢化方法
EP1756084B1 (de) 2004-06-04 2008-11-26 Arena Pharmaceuticals, Inc. Substituierte aryl- und heteroarylderivate als modulatoren des stoffwechsels und für die prophylaxe und behandlung von damit in zusammenhang stehenden erkrankungen
WO2005123677A1 (en) 2004-06-16 2005-12-29 Actelion Pharmaceuticals Ltd 4-carbonyl substituted 1,1,2-trimethyl-1a,4,5,5a-tetrahydro-1h-4-aza-cyclopropa'a!pentalene derivatives as agonists for the g-protein-coupled receptor s1p1/edg1 and immunosuppressive agents
US8039674B2 (en) 2004-06-23 2011-10-18 Ono Pharmaceutical Co., Ltd. Compound having S1P receptor binding potency and use thereof
KR101178318B1 (ko) 2004-07-16 2012-08-29 교린 세이야꾸 가부시키 가이샤 효과적인 의약의 사용법 및 부작용 발현의 방어에 관한방법
JP2007284350A (ja) 2004-07-27 2007-11-01 Takeda Chem Ind Ltd 糖尿病治療剤
TW200611687A (en) 2004-07-29 2006-04-16 Sankyo Co Pharmaceutical compositions used for immunosuppressant
WO2006010379A1 (en) 2004-07-29 2006-02-02 Actelion Pharmaceuticals Ltd. Novel thiophene derivatives as immunosuppressive agents
EP1798226A4 (de) 2004-08-04 2009-06-17 Taisho Pharmaceutical Co Ltd Triazolderivat
US20060223866A1 (en) 2004-08-13 2006-10-05 Praecis Pharmaceuticals, Inc. Methods and compositions for modulating sphingosine-1-phosphate (S1P) receptor activity
KR20070058455A (ko) 2004-08-13 2007-06-08 프래시스 파마슈티컬즈 인코포레이티드 스핑고신-1-포스페이트(s1p) 수용체 활성을 조절하기위한 방법 및 조성물
BRPI0515596A (pt) 2004-09-23 2008-07-29 Wyeth Corp composição farmacêutica usada para o tratamento e/ou prevenção de hcv, e, métodos de inibir rna polimerase ns5b de hepatite c de tratar ou prevenir infecção por hepatite c em um mamìfero
AU2005295080A1 (en) 2004-10-12 2006-04-20 Transition Therapeutics Inc. Compounds and methods of treating insulin resistance and cardiomyopathy
AU2005299851B2 (en) 2004-10-22 2011-03-17 Merck Sharp & Dohme Corp. 2-(aryl)azacyclylmethyl carboxylates, sulfonates, phosphonates, phosphinates and heterocycles as S1P receptor agonists
SI1650186T1 (sl) 2004-10-22 2008-12-31 Bioprojet Soc Civ Novi derivati dikarboksilne kisline
EP1844043A2 (de) 2004-11-18 2007-10-17 The Institutes for Pharmaceutical Discovery, LLC Heterocyclisch substituierte carboxylsäuren zur behandlung von diabetes
WO2006057448A1 (ja) 2004-11-26 2006-06-01 Takeda Pharmaceutical Company Limited アリールアルカン酸誘導体
MX2007006706A (es) 2004-12-06 2007-10-18 Univ Virginia Analogos de esfingosina 1-fosfato amida de arilo.
AU2005314938B2 (en) 2004-12-13 2011-06-09 Ono Pharmaceutical Co., Ltd. Aminocarboxylic acid derivative and medicinal use thereof
KR20070086007A (ko) 2004-12-13 2007-08-27 다이이찌 산쿄 가부시키가이샤 당뇨병의 치료를 위한 의약 조성물
MY148521A (en) 2005-01-10 2013-04-30 Arena Pharm Inc Substituted pyridinyl and pyrimidinyl derivatives as modulators of metabolism and the treatment of disorders related thereto
DOP2006000010A (es) 2005-01-10 2006-07-31 Arena Pharm Inc Procedimiento para preparar eteres aromáticos
DOP2006000008A (es) 2005-01-10 2006-08-31 Arena Pharm Inc Terapia combinada para el tratamiento de la diabetes y afecciones relacionadas y para el tratamiento de afecciones que mejoran mediante un incremento de la concentración sanguínea de glp-1
DOP2006000009A (es) 2005-01-13 2006-08-15 Arena Pharm Inc Procedimiento para preparar eteres de pirazolo [3,4-d] pirimidina
DE112005003337T5 (de) 2005-01-25 2008-02-21 Merck Patent Gmbh Mesogene Verbindungen, Flüssigkristallmedium und Flüssigkristallanzeige
EP1685841A1 (de) 2005-01-31 2006-08-02 Bayer Health Care Aktiengesellschaft Prophylaxe und Behandlung von thrombolischen Erkrankungen
AU2006214314B2 (en) 2005-02-14 2012-02-09 University Of Virginia Patent Foundation Sphingosine 1- phos phate agonists comprising cycloalkanes and 5 -membered heterocycles substituted by amino and phenyl groups
ES2365747T3 (es) 2005-03-23 2011-10-10 Actelion Pharmaceuticals Ltd. Derivados de benzo[c]tiofeno hidrogenados como inmunomoduladores.
CN101180050B (zh) 2005-03-23 2011-07-27 埃科特莱茵药品有限公司 作为鞘氨醇-1-磷酸盐-1受体激动剂的新颖噻吩衍生物
AU2006226020A1 (en) 2005-03-23 2006-09-28 Actelion Pharmaceuticals Ltd. Novel thiophene derivatives as sphingosine-l-phosphate-1 receptor agonists
BRPI0612028A2 (pt) 2005-06-08 2010-10-13 Novartis Ag oxadiazóis ou isodiazóis policìclicos e uso dos mesmos como ligantes de receptor s1p
JP2008546758A (ja) 2005-06-24 2008-12-25 アクテリオン ファーマシューティカルズ リミテッド 新規チオフェン誘導体
TWI418350B (zh) 2005-06-24 2013-12-11 Sankyo Co 含有ppar調節劑之醫藥組成物的用途
MX2007016545A (es) 2005-06-30 2008-02-21 Prosidion Ltd Agonistas del receptor acoplado a la proteina g.
US20070060573A1 (en) 2005-08-10 2007-03-15 Lars Wortmann Acyltryptophanols
AU2006283175A1 (en) 2005-08-23 2007-03-01 Irm Llc Immunosuppressant compounds and compositions
US20100160359A1 (en) 2005-09-16 2010-06-24 Arena Pharmaceuticals, Inc. Modulators of Metabolism and the Treatment of Disorders Related Thereto
JPWO2007037196A1 (ja) 2005-09-29 2009-04-09 山本化成株式会社 インドリン系化合物及びその製造方法
AU2006300485B2 (en) 2005-10-07 2011-08-25 Kyorin Pharmaceutical Co., Ltd. Therapeutic agent for liver disease containing 2-amino-1,3-propanediol derivative as active ingredient and therapeutic method for liver disease
AR057894A1 (es) 2005-11-23 2007-12-26 Actelion Pharmaceuticals Ltd Derivados de tiofeno
EP1965807A4 (de) 2005-11-23 2010-10-27 Epix Delaware Inc S1p-rezeptor-modulierende verbindungen und ihre verwendung
TWI404706B (zh) 2006-01-11 2013-08-11 Actelion Pharmaceuticals Ltd 新穎噻吩衍生物
TW200736234A (en) 2006-01-17 2007-10-01 Astrazeneca Ab Chemical compounds
BRPI0621226A2 (pt) 2006-01-19 2012-07-10 Orchid Reseach Lab Ltd compostos heterociclos
ATE447568T1 (de) 2006-01-24 2009-11-15 Actelion Pharmaceuticals Ltd Neue pyrinderivate
GB0601744D0 (en) 2006-01-27 2006-03-08 Novartis Ag Organic compounds
TWI389683B (zh) 2006-02-06 2013-03-21 Kyorin Seiyaku Kk A therapeutic agent for an inflammatory bowel disease or an inflammatory bowel disease treatment using a 2-amino-1,3-propanediol derivative as an active ingredient
TW200806611A (en) 2006-02-09 2008-02-01 Daiichi Seiyaku Co Novel amidopropionic acid derivatives and medicine containing the same
CA2641718A1 (en) 2006-02-09 2007-08-16 University Of Virginia Patent Foundation Bicyclic sphingosine 1-phosphate analogs
US20070191371A1 (en) 2006-02-14 2007-08-16 Kalypsys, Inc. Heterocyclic modulators of ppar
CN101460458A (zh) 2006-02-15 2009-06-17 阿勒根公司 具有1-磷酸-鞘氨醇(s1p)受体拮抗剂生物活性的带芳基或者杂芳基基团的吲哚-3-羧酸的酰胺、酯、硫代酰胺和硫羟酸酯化合物
AU2007214434B2 (en) 2006-02-15 2012-06-14 Allergan, Inc. Indole-3-carboxylic acid amide, ester, thioamide and thiol ester compounds bearing aryl or heteroaryl groups having sphingosine-1-phosphate (S1P) receptor antagonist biological activity
EP1991535A1 (de) 2006-02-21 2008-11-19 University Of Virginia Patent Foundation Phenyl-cycloalkyl und phenyl-heterocyclische derivate als sip-rezeptoragonisten
US7649098B2 (en) 2006-02-24 2010-01-19 Lexicon Pharmaceuticals, Inc. Imidazole-based compounds, compositions comprising them and methods of their use
AU2007225208A1 (en) 2006-03-14 2007-09-20 Amgen Inc. Bicyclic carboxylic acid derivatives useful for treating metabolic disorders
AU2007227274A1 (en) 2006-03-21 2007-09-27 Epix Delaware, Inc. S1P receptor modulating compounds
JP2007262009A (ja) 2006-03-29 2007-10-11 Dai Ichi Seiyaku Co Ltd ヘテロアリール低級カルボン酸誘導体
MX356221B (es) 2006-04-03 2018-05-18 Astellas Pharma Inc Heterocompuesto.
GB0607389D0 (en) 2006-04-12 2006-05-24 Novartis Ag Organic compounds
JPWO2007129745A1 (ja) 2006-05-09 2009-09-17 第一三共株式会社 ヘテロアリールアミド低級カルボン酸誘導体
CN101490046A (zh) 2006-05-09 2009-07-22 辉瑞产品公司 环烷基氨基酸衍生物及其药物组合物
WO2007129473A1 (ja) 2006-05-09 2007-11-15 Daiichi Sankyo Company, Limited 二環性アリール誘導体
TW200811140A (en) 2006-07-06 2008-03-01 Arena Pharm Inc Modulators of metabolism and the treatment of disorders related thereto
TW200811147A (en) 2006-07-06 2008-03-01 Arena Pharm Inc Modulators of metabolism and the treatment of disorders related thereto
TW200823182A (en) 2006-08-01 2008-06-01 Praecis Pharm Inc Chemical compounds
US20080070866A1 (en) 2006-08-01 2008-03-20 Praecis Pharmaceuticals Incorporated Chemical compounds
JP2009545630A (ja) 2006-08-04 2009-12-24 プリーシス・ファーマシューティカルズ・インコーポレイテッド 化合物
US8232319B2 (en) 2006-08-08 2012-07-31 Kyorin Pharmaceutical Co., Ltd. Amino phosphate derivative and S1P receptor modulator having same as an active ingredient
JP2010501555A (ja) 2006-08-24 2010-01-21 プリーシス・ファーマシューティカルズ・インコーポレイテッド S1p−1受容体アゴニスト
JP2009269819A (ja) 2006-08-25 2009-11-19 Asahi Kasei Pharma Kk アミン化合物
JP5253401B2 (ja) 2006-09-07 2013-07-31 アクテリオン ファーマシューティカルズ リミテッド 免疫調節薬としてのピリジン−4−イル誘導体
WO2008030843A1 (en) 2006-09-07 2008-03-13 Allergan, Inc. Heteroaromatic compounds having sphingosine-1-phosphate (s1p) receptor agonist and/or antagonist biological activity
TWI408139B (zh) 2006-09-07 2013-09-11 Actelion Pharmaceuticals Ltd 新穎噻吩衍生物
CA2662091A1 (en) 2006-09-08 2008-03-13 Novartis Ag N-biaryl (hetero) arylsulphonamide derivatives useful in the treatment of diseases mediated by lymphocytes interactions
WO2008035239A1 (en) 2006-09-21 2008-03-27 Actelion Pharmaceuticals Ltd Phenyl derivatives and their use as immunomodulators
US8101634B2 (en) 2006-12-06 2012-01-24 Glaxosmithkline Llc Bicyclic compounds and use as antidiabetics
WO2008076356A1 (en) 2006-12-15 2008-06-26 Abbott Laboratories Novel oxadiazole compounds
JO2701B1 (en) 2006-12-21 2013-03-03 جلاكسو جروب ليميتد Vehicles
GB0625647D0 (en) * 2006-12-21 2007-01-31 Glaxo Group Ltd Compounds
UY30829A1 (es) 2006-12-21 2008-07-31 Abbott Lab Compuestos agonistas y antagonistas del receptor de esfinfosina-1-fosfato
GB0625648D0 (en) 2006-12-21 2007-01-31 Glaxo Group Ltd Compounds
CN101668741A (zh) 2007-01-11 2010-03-10 阿勒根公司 具有鞘氨醇-1-磷酸(s1p)受体拮抗剂生物学活性的6-取代吲哚-3-羧酸酰胺化合物
WO2008091967A1 (en) 2007-01-26 2008-07-31 Smithkline Beecham Corporation Chemical compounds
WO2008097819A2 (en) 2007-02-05 2008-08-14 Smithkline Beecham Corporation Chemical compounds
NZ580454A (en) 2007-03-16 2011-05-27 Actelion Pharmaceuticals Ltd Amino- pyridine derivatives as s1p1 /edg1 receptor agonists
AU2007227278B2 (en) 2007-03-21 2014-09-11 Epix Pharmaceuticals, Inc. SIP receptor modulating compounds and use thereof
ATE500252T1 (de) 2007-04-19 2011-03-15 Glaxo Group Ltd Oxadiazolsubstituierte indazolderivate zur verwendung als sphingosin-1-phosphat (s1p) - agonisten
EP2014653A1 (de) 2007-06-15 2009-01-14 Bioprojet Neue Dicarbonsäurederivate als S1P1 Rezeptor Agonisten
US20090069288A1 (en) 2007-07-16 2009-03-12 Breinlinger Eric C Novel therapeutic compounds
WO2009019506A1 (en) 2007-08-03 2009-02-12 Astrazeneca Ab Heterocyclyc sulfonamides having edg-1 antagonistic activity
US8399448B2 (en) 2007-08-08 2013-03-19 Merck Serono Sa 6-amino-pyrimidine-4-carboxamide derivatives and related compounds which bind to the sphingosine 1-phosphate (S1P) receptor for the treatment of multiple sclerosis
AU2008302570B2 (en) 2007-09-20 2012-05-31 Irm Llc Compounds and compositions as modulators of GPR119 activity
UY31468A1 (es) 2007-11-15 2009-07-17 Derivados bis-(sulfonilamino) en terapia 065
US8637704B2 (en) 2007-11-28 2014-01-28 Synta Pharmaceuticals Corp. Polymorphs of N-malonyl-bis(N′-methyl-N′-thiobenzoylhydrazide)
JP2011506600A (ja) * 2007-12-18 2011-03-03 アリーナ ファーマシューティカルズ, インコーポレイテッド 自己免疫性障害および炎症性障害の治療において有用なテトラヒドロシクロペンタ[b]インドール−3−イルカルボン酸誘導体
AU2009206733A1 (en) 2008-01-25 2009-07-30 Arena Pharmaceuticals, Inc. Dihydro- 1H- pyrrolo [1,2-a] indol-1-yl carboxylic derivatives which act as S1P1 agonists
AR070398A1 (es) 2008-02-22 2010-03-31 Gruenenthal Chemie Derivados sustituidos de indol
WO2009108924A2 (en) 2008-02-28 2009-09-03 Solar Roofing Systems, Inc. Photovoltaic roofing tile with fire suppression
ES2617628T5 (es) 2008-03-17 2020-06-01 Actelion Pharmaceuticals Ltd Régimen de dosificación para un agonista selectivo del receptor de S1P1
ES2813368T3 (es) 2008-05-14 2021-03-23 Scripps Research Inst Nuevos moduladores de receptores de fosfato de esfingosina
WO2009151621A1 (en) 2008-06-13 2009-12-17 Arena Pharmaceuticals, Inc. Substituted (1, 2, 4-0xadiaz0l-3-yl) indolin-1-yl carboxylic acid derivatives useful as s1p1 agonists
WO2009151626A1 (en) 2008-06-13 2009-12-17 Arena Pharmaceuticals, Inc. Substituted (1, 2, 4-0xadiaz0l-3-yl) indolin-1-yl carboxylic acid derivatives useful as s1p1 agonists
KR20180023049A (ko) * 2008-07-23 2018-03-06 아레나 파마슈티칼스, 인크. 자가면역성 및 염증성의 장애의 치료에 유용한 치환된 1,2,3,4-테트라히드로시클로펜타[b]인돌-3-일)아세트산 유도체
EP2342205B1 (de) 2008-08-27 2016-04-20 Arena Pharmaceuticals, Inc. Substituierte tricyclische carbonsäurederivative als s1p1 rezeptor agonisten für die behandlung von entzündlichen und autoimmunerkrankungen
AR074825A1 (es) 2008-12-22 2011-02-16 Novartis Ag Regimen de dosificacion de un agonista de los receptores de s1p, metodo de tratamiento y kit
LT3409274T (lt) 2008-12-22 2020-02-10 Novartis Ag Dozavimo schema s1p receptoriaus agonistui
JP2012517446A (ja) 2009-02-10 2012-08-02 アボット・ラボラトリーズ S1p5受容体の作動薬および拮抗薬ならびにそれらの使用方法
WO2011005290A1 (en) 2009-06-23 2011-01-13 Arena Pharmaceuticals, Inc. Disubstituted oxadiazole derivatives useful in the treatment of autoimmune and inflammatory disorders
WO2011005295A1 (en) 2009-06-24 2011-01-13 Arena Pharmaceuticals, Inc. Modulators of the sphingosine-1-phosphate (s1p) receptor useful for the treatment of disorders related thereto
WO2011059784A1 (en) 2009-10-29 2011-05-19 Bristol-Myers Squibb Company Tricyclic heterocyclic compounds
US8299059B2 (en) 2009-10-30 2012-10-30 Eli Lilly And Company Crystalline compound and a process for its preparation
US8440693B2 (en) 2009-12-22 2013-05-14 Novartis Ag Substituted isoquinolinones and quinazolinones
EP2470529B1 (de) 2009-12-30 2014-11-19 Medichem, S.A. Pharmazeutische verwendung eines 1-(1h-1,2,4-triazol-1-yl)butan-2-ol derivats und verwendung eines 1-(1h-1,2,4-triazol-1-yl)butan-2-ol derivats mit nicht definierter kristallform zur herstellung besagten 1-(1h-1,2,4-triahol-1-yl)butan-2-ol derivats
EP4148045A1 (de) * 2010-01-27 2023-03-15 Arena Pharmaceuticals, Inc. Zwischenverbindungen zur herstellung von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)essigsäure und salzen davon
SG10201501575VA (en) 2010-03-03 2015-04-29 Arena Pharm Inc Processes for the preparation of s1p1 receptor modulators and crystalline forms thereof
BR112012025592A2 (pt) 2010-04-06 2019-09-24 Arena Pharm Inc moduladores do receptor de gpr119 e o tratamento de distúrbios relacionados com os mesmos
WO2012015758A2 (en) 2010-07-30 2012-02-02 Saint Louis University Methods of treating pain
JP5941916B2 (ja) 2010-09-22 2016-06-29 アリーナ ファーマシューティカルズ, インコーポレイテッド Gpr119レセプターのモジュレーターおよびそれに関連する障害の処置
BR112013019683B1 (pt) 2011-02-07 2022-04-19 Biogen Inc Compostos que modulam s1p, composição farmacêutica, e uso dos mesmos
WO2012145603A1 (en) 2011-04-22 2012-10-26 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
BR112014018485A8 (pt) 2012-02-03 2017-07-11 Teva Pharma Uso de laquinimod para tratar pacientes com a doença de crohn que falharam com a primeira linha de terapia anti-tnf
US20160038506A1 (en) 2012-02-14 2016-02-11 Repros Therapeutics Inc. Selective Estrogen Receptor Modulators With Short Half-Lives and Uses Thereof
US8962888B2 (en) 2012-12-03 2015-02-24 Physical Sciences, Inc. Forming spherical crystal habit
JP5589110B1 (ja) 2013-03-08 2014-09-10 株式会社ポーラファルマ 晶癖を有する結晶及び該結晶を有効成分として含有する医薬組成物
JP6895378B2 (ja) 2015-01-06 2021-06-30 アリーナ ファーマシューティカルズ, インコーポレイテッド S1p1受容体に関連する状態の処置方法
EP3939965A1 (de) 2015-06-22 2022-01-19 Arena Pharmaceuticals, Inc. Kristallines l-arginin-salz von (r)-2-(7-(4-cyclopentyl-3-(trifluormethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta[b]indol-3-yl)essigsäure zur verwendung in s1p1-rezeptor-assoziierten erkrankungen
CN110520124A (zh) 2017-02-16 2019-11-29 艾尼纳制药公司 用于治疗原发性胆汁性胆管炎的化合物和方法
WO2018151834A1 (en) 2017-02-16 2018-08-23 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of inflammatory bowel disease with extra-intestinal manifestations
CA3102136A1 (en) 2018-06-06 2019-12-12 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the s1p1 receptor
WO2020072824A1 (en) 2018-10-03 2020-04-09 Arena Pharmaceuticals, Inc. Methods for the treatment of scleroderma
CN113226307A (zh) 2018-11-30 2021-08-06 艾尼纳制药公司 治疗与s1p1受体有关的病况的方法
EP3908276A1 (de) 2019-01-08 2021-11-17 Arena Pharmaceuticals, Inc. Verfahren zur behandlung von zuständen im zusammenhang mit dem s1p1-rezeptor

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
None *
See also references of WO2011094008A1 *

Also Published As

Publication number Publication date
US9447041B2 (en) 2016-09-20
JP2013518106A (ja) 2013-05-20
EP3378854A1 (de) 2018-09-26
CA2786994C (en) 2018-01-16
CN103221391A (zh) 2013-07-24
SG10201906876PA (en) 2019-09-27
CN108558740B (zh) 2021-10-19
WO2011094008A1 (en) 2011-08-04
US20120295947A1 (en) 2012-11-22
US20160016904A1 (en) 2016-01-21
US8853419B2 (en) 2014-10-07
CN103221391B (zh) 2018-07-06
JP2015154782A (ja) 2015-08-27
US11674163B2 (en) 2023-06-13
SG10201500639TA (en) 2015-03-30
CA2786994A1 (en) 2011-08-04
SG182610A1 (en) 2012-08-30
US9175320B2 (en) 2015-11-03
EP4148045A1 (de) 2023-03-15
US20170159088A1 (en) 2017-06-08
JP5856980B2 (ja) 2016-02-10
EP3378854B1 (de) 2022-12-21
US20220259627A1 (en) 2022-08-18
CN108558740A (zh) 2018-09-21
ES2937386T3 (es) 2023-03-28
US11149292B2 (en) 2021-10-19
US20140357690A1 (en) 2014-12-04

Similar Documents

Publication Publication Date Title
US11674163B2 (en) Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
US20200407316A1 (en) SUBSTITUTED (1,2,3,4-TETRAHYDROCYCLOPENTA[b]INDOL-3-YL)ACETIC ACID DERIVATIVES USEFUL IN THE TREATMENT OF AUTOIMMUNE AND INFLAMMATORY DISORDERS
CA2733671C (en) Substituted tricyclic acid derivatives as s1p1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US9085581B2 (en) Processes for the preparation of S1P1 receptor modulators and crystalline forms thereof
EP3847158A1 (de) Verbindungen zur verwendung bei der behandlung von autoimmun- und entzündlichen erkrankungen

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20120824

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ARENA PHARMACEUTICALS, INC.

17Q First examination report despatched

Effective date: 20130723

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1177733

Country of ref document: HK

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20160421

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20170105

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTC Intention to grant announced (deleted)
INTG Intention to grant announced

Effective date: 20171005

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20180216